US9770410B2 - Methods and compositions for CNS delivery of arylsulfatase A - Google Patents

Methods and compositions for CNS delivery of arylsulfatase A Download PDF

Info

Publication number
US9770410B2
US9770410B2 US13/168,963 US201113168963A US9770410B2 US 9770410 B2 US9770410 B2 US 9770410B2 US 201113168963 A US201113168963 A US 201113168963A US 9770410 B2 US9770410 B2 US 9770410B2
Authority
US
United States
Prior art keywords
protein
brain
formulation
rhasa
asa
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US13/168,963
Other languages
English (en)
Other versions
US20120009171A1 (en
Inventor
Nazila Salamat-Miller
Katherine Taylor
Paul Campolieto
Zahra Shahrokh
Jing Pan
Lawrence Charnas
Teresa Leah Wright
Pericles Calias
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Takeda Pharmaceutical Co Ltd
Original Assignee
Shire Human Genetics Therapies Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shire Human Genetics Therapies Inc filed Critical Shire Human Genetics Therapies Inc
Priority to US13/168,963 priority Critical patent/US9770410B2/en
Assigned to SHIRE HUMAN GENETIC THERAPIES, INC. reassignment SHIRE HUMAN GENETIC THERAPIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CALIAS, PERICLES, WRIGHT, TERESA LEAH, CAMPOLIETO, PAUL, TAYLOR, KATHERINE, CHARNAS, LAWRENCE, PAN, JING, SALAMAT-MILLER, NAZILA, SHAHROKH, ZAHRA
Publication of US20120009171A1 publication Critical patent/US20120009171A1/en
Priority to US15/684,568 priority patent/US10646554B2/en
Application granted granted Critical
Publication of US9770410B2 publication Critical patent/US9770410B2/en
Priority to US16/828,731 priority patent/US20200405825A1/en
Assigned to TAKEDA PHARMACEUTICAL COMPANY LIMITED reassignment TAKEDA PHARMACEUTICAL COMPANY LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SHIRE HUMAN GENETIC THERAPIES, INC.
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0085Brain, e.g. brain implants; Spinal cord
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/465Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/54Mixtures of enzymes or proenzymes covered by more than a single one of groups A61K38/44 - A61K38/46 or A61K38/51 - A61K38/53
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/06Sulfuric ester hydrolases (3.1.6)
    • C12Y301/06013Iduronate-2-sulfatase (3.1.6.13)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner

Definitions

  • Enzyme replacement therapy involves the systemic administration of natural or recombinantly-derived proteins and/or enzymes to a subject.
  • Approved therapies are typically administered to subjects intravenously and are generally effective in treating the somatic symptoms of the underlying enzyme deficiency.
  • the intravenously administered protein and/or enzyme into the cells and tissues of the central nervous system (CNS)
  • the treatment of diseases having a CNS etiology has been especially challenging because the intravenously administered proteins and/or enzymes do not adequately cross the blood-brain barrier (BBB).
  • the blood-brain barrier is a structural system comprised of endothelial cells that functions to protect the central nervous system (CNS) from deleterious substances in the blood stream, such as bacteria, macromolecules (e.g., proteins) and other hydrophilic molecules, by limiting the diffusion of such substances across the BBB and into the underlying cerebrospinal fluid (CSF) and CNS.
  • CNS central nervous system
  • Intrathecal (IT) injection or the administration of proteins to the cerebrospinal fluid (CSF) has also been attempted but has not yet yielded therapeutic success.
  • a major challenge in this treatment has been the tendency of the active agent to bind the ependymal lining of the ventricle very tightly which prevented subsequent diffusion.
  • GAGs glycosaminoglycans
  • the present invention provides an effective and less invasive approach for direct delivery of therapeutic agents to the central nervous system (CNS).
  • CNS central nervous system
  • a replacement enzyme e.g., arylsulfatase A (ASA)
  • ASA arylsulfatase A
  • MLD lysosomal storage disease
  • CSF cerebrospinal fluid
  • the present inventors have demonstrated that such high protein concentration delivery can be done using simple saline or buffer-based formulations and without inducing substantial adverse effects, such as severe immune response, in the subject. Therefore, the present invention provides a highly efficient, clinically desirable and patient-friendly approach for direct CNS delivery for the treatment various diseases and disorders that have CNS components, in particular, lysosomal storage diseases.
  • the present invention represents a significant advancement in the field of CNS targeting and enzyme replacement therapy.
  • stable formulations for effective intrathecal (IT) administration of an arylsulfatase A (ASA) protein As described in detail below, the present inventors have successfully developed stable formulations for effective intrathecal (IT) administration of an arylsulfatase A (ASA) protein. It is contemplated, however, that various stable formulations described herein are generally suitable for CNS delivery of therapeutic agents, including various other lysosomal enzymes. Indeed, stable formulations according to the present invention can be used for CNS delivery via various techniques and routes including, but not limited to, intraparenchymal, intracerebral, intravetricular cerebral (ICV), intrathecal (e.g., IT-Lumbar, IT-cisterna magna) administrations and any other techniques and routes for injection directly or indirectly to the CNS and/or CSF.
  • ICV intravetricular cerebral
  • intrathecal e.g., IT-Lumbar, IT-cisterna magna
  • a replacement enzyme can be a synthetic, recombinant, gene-activated or natural enzyme.
  • the present invention includes a stable formulation for direct CNS intrathecal administration comprising an arylsulfatase A (ASA) protein, salt, and a polysorbate surfactant.
  • ASA arylsulfatase A
  • the ASA protein is present at a concentration ranging from approximately 1-300 mg/ml (e.g., 1-250 mg/ml, 1-200 mg/ml, 1-150 mg/ml, 1-100 mg/ml, 1-50 mg/ml).
  • the ASA protein is present at or up to a concentration selected from 2 mg/ml, 3 mg/ml, 4 mg/ml, 5 mg/ml, 10 mg/ml, 15 mg/ml, 20 mg/ml, 25 mg/ml, 30 mg/ml, 35 mg/ml, 40 mg/ml, 45 mg/ml, 50 mg/ml, 60 mg/ml, 70 mg/ml, 80 mg/ml, 90 mg/ml, 100 mg/ml, 150 mg/ml, 200 mg/ml, 250 mg/ml, or 300 mg/ml.
  • the present invention includes a stable formulation of any of the embodiments described herein, wherein the ASA protein comprises an amino acid sequence of SEQ ID NO:1.
  • the ASA protein comprises an amino acid sequence at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 98% identical to SEQ ID NO:1.
  • the stable formulation of any of the embodiments described herein includes a salt.
  • the salt is NaCl.
  • the NaCl is present as a concentration ranging from approximately 0-300 mM (e.g., 0-250 mM, 0-200 mM, 0-150 mM, 0-100 mM, 0-75 mM, 0-50 mM, or 0-30 mM). In some embodiments, the NaCl is present at a concentration ranging from approximately 137-154 mM. In some embodiments, the NaCl is present at a concentration of approximately 154 mM.
  • the present invention includes a stable formulation of any of the embodiments described herein, wherein the polysorbate surfactant is selected from the group consisting of polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80 and combination thereof.
  • the polysorbate surfactant is polysorbate 20.
  • the polysorbate 20 is present at a concentration ranging approximately 0-0.02%. In some embodiments, the polysorbate 20 is present at a concentration of approximately 0.005%.
  • the present invention includes a stable formulation of any of the embodiments described herein, wherein the formulation further comprises a buffering agent.
  • the buffering agent is selected from the group consisting of phosphate, acetate, histidine, sccinate, Tris, and combinations thereof.
  • the buffering agent is phosphate.
  • the phosphate is present at a concentration no greater than 50 mM (e.g., no greater than 45 mM, 40 mM, 35 mM, 30 mM, 25 mM, 20 mM, 15 mM, 10 mM, or 5 mM).
  • the phosphate is present at a concentration no greater than 20 mM.
  • the invention includes a stable formulation of any of the embodiments described herein, wherein the formulation has a pH of approximately 3-8 (e.g., approximately 4-7.5, 5-8, 5-7.5, 5-6.5, 5-7.0, 5.5-8.0, 5.5-7.7, 5.5-6.5, 6-7.5, or 6-7.0).
  • the formulation has a pH of approximately 5.5-6.5 (e.g., 5.5, 6.0, 6.1, 6.2, 6.3, 6.4, or 6.5).
  • the formulation has a pH of approximately 6.0.
  • the present invention includes stable formulations of any of the embodiments described herein, wherein the formulation is a liquid formulation. In various embodiments, the present invention includes stable formulation of any of the embodiments described herein, wherein the formulation is formulated as lyophilized dry powder.
  • the present invention includes a stable formulation for intrathecal administration comprising an arylsulfatase A (ASA) protein at a concentration ranging from approximately 1-300 mg/ml, NaCl at a concentration of approximately 154 mM, polysorbate 20 at a concentration of approximately 0.005%, and a pH of approximately 6.0.
  • ASA arylsulfatase A
  • the ASA protein is at a concentration of approximately 10 mg/ml.
  • the ASA protein is at a concentration of approximately 30 mg/ml, 40 mg/ml, 50 mg/ml, 100 mg/ml, 150 mg/ml, 200 mg/ml, 250 mg/ml, or 300 mg/ml.
  • the present invention includes a container comprising a single dosage form of a stable formulation in various embodiments described herein.
  • the container is selected from an ampule, a vial, a bottle, a cartridge, a reservoir, a lyo-ject, or a pre-filled syringe.
  • the container is a pre-filled syringe.
  • the pre-filled syringe is selected from borosilicate glass syringes with baked silicone coating, borosilicate glass syringes with sprayed silicone, or plastic resin syringes without silicone.
  • the stable formulation is present in a volume of less than about 50 mL (e.g., less than about 45 ml, 40 ml, 35 ml, 30 ml, 25 ml, 20 ml, 15 ml, 10 ml, 5 ml, 4 ml, 3 ml, 2.5 ml, 2.0 ml, 1.5 ml, 1.0 ml, or 0.5 ml). In some embodiments, the stable formulation is present in a volume of less than about 3.0 mL.
  • the present invention includes methods of treating Metachromatic Leukodystrophy Disease including the step of administering intrathecally to a subject in need of treatment a formulation according to any of the embodiments described herein.
  • the present invention includes a method of treating Metachromatic Leukodystrophy Disease including a step of administering intrathecally to a subject in need of treatment a formulation comprising an arylsulfatase A (ASA) protein at a concentration ranging from approximately 1-300 mg/ml, NaCl at a concentration of approximately 154 mM, polysorbate 20 at a concentration of approximately 0.005%, and a pH of approximately 6.
  • ASA arylsulfatase A
  • the intrathecal administration results in no substantial adverse effects (e.g., severe immune response) in the subject. In some embodiments, the intrathecal administration results in no substantial adaptive T cell-mediated immune response in the subject.
  • the intrathecal administration of the formulation results in delivery of the arylsulfatase A protein to various target tissues in the brain, the spinal cord, and/or peripheral organs. In some embodiments, the intrathecal administration of the formulation results in delivery of the arylsulfatase A protein to target brain tissues.
  • the brain target tissues comprise white matter and/or neurons in the gray matter.
  • the arylsulfatase A protein is delivered to neurons, glial cells, perivascular cells and/or meningeal cells. In some embodiments, the arylsulfatase A protein is further delivered to the neurons in the spinal cord.
  • the intrathecal administration of the formulation further results in systemic delivery of the ASA protein in peripheral target tissues.
  • the peripheral target tissues are selected from liver, kidney, spleen and/or heart.
  • the intrathecal administration of the formulation results in lysosomal localization in brain target tissues, spinal cord neurons and/or peripheral target tissues. In some embodiments, the intrathecal administration of the formulation results in reduction of sulfatide storage in the brain target tissues, spinal cord neurons and/or peripheral target tissues. In some embodiments, the sulfatide storage is reduced by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 1-fold, 1.5-fold, or 2-fold as compared to a control (e.g., the pre-treatment GAG storage in the subject).
  • a control e.g., the pre-treatment GAG storage in the subject.
  • the intrathecal administration of the formulation results in reduced vacuolization in neurons (e.g., by at least 20%, 40%, 50%, 60%, 80%, 90%, 1-fold, 1.5-fold, or 2-fold as compared to a control).
  • the neurons comprises Purkinje cells.
  • the intrathecal administration of the formulation results in increased ASA enzymatic activity in the brain target tissues, spinal cord neurons and/or peripheral target tissues.
  • the ASA enzymatic activity is increased by at least 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold or 10-fold as compared to a control (e.g., the pre-treatment endogenous enzymatic activity in the subject).
  • the increased ASA enzymatic activity is at least approximately 10 nmol/hr/mg, 20 nmol/hr/mg, 40 nmol/hr/mg, 50 nmol/hr/mg, 60 nmol/hr/mg, 70 nmol/hr/mg, 80 nmol/hr/mg, 90 nmol/hr/mg, 100 nmol/hr/mg, 150 nmol/hr/mg, 200 nmol/hr/mg, 250 nmol/hr/mg, 300 nmol/hr/mg, 350 nmol/hr/mg, 400 nmol/hr/mg, 450 nmol/hr/mg, 500 nmol/hr/mg, 550 nmol/hr/mg or 600 nmol/hr/mg.
  • the ASA enzymatic activity is increased in the lumbar region.
  • the increased ASA enzymatic activity in the lumbar region is at least approximately 2000 nmol/hr/mg, 3000 nmol/hr/mg, 4000 nmol/hr/mg, 5000 nmol/hr/mg, 6000 nmol/hr/mg, 7000 nmol/hr/mg, 8000 nmol/hr/mg, 9000 nmol/hr/mg, or 10,000 nmol/hr/mg.
  • the intrathecal administration of the formulation results in reduced intensity, severity, or frequency, or delayed onset of at least one symptom or feature of MLD.
  • the at least one symptom or feature of the MLD is cognitive impairment; white matter lesions; dilated perivascular spaces in the brain parenchyma, ganglia, corpus callosum, and/or brainstem; atrophy; and/or ventriculomegaly.
  • the intrathecal administration takes place once every two weeks. In some embodiments, the intrathecal administration takes place once every month. In some embodiments, the intrathecal administration takes place once every two months. In some embodiments, the intrathecal administration is used in conjunction with intravenous administration. In some embodiments, the intravenous administration is no more frequent than once every week. In some embodiments, the intravenous administration is no more frequent than once every two weeks. In some embodiments, the intravenous administration is no more frequent than once every month. In some embodiments, the intravenous administration is no more frequent than once every two months. In certain embodiments, the intraveneous administration is more frequent than monthly administration, such as twice weekly, weekly, every other week, or twice monthly.
  • intraveneous and intrathecal administrations are performed on the same day. In some embodiments, the intraveneous and intrathecal administrations are not performed within a certain amount of time of each other, such as not within at least 2 days, within at least 3 days, within at least 4 days, within at least 5 days, within at least 6 days, within at least 7 days, or within at least one week. In some embodiments, intraveneous and intrathecal administrations are performed on an alternating schedule, such as alternating administrations weekly, every other week, twice monthly, or monthly.
  • an intrathecal administration replaces an intravenous administration in an administration schedule, such as in a schedule of intraveneous administration weekly, every other week, twice monthly, or monthly, every third or fourth or fifth administration in that schedule can be replaced with an intrathecal administration in place of an intraveneous administration.
  • intraveneous and intrathecal administrations are performed sequentially, such as performing intraveneous administrations first (e.g., weekly, every other week, twice monthly, or monthly dosing for two weeks, a month, two months, three months, four months, five months, six months, a year or more) followed by IT administrations (e.g., weekly, every other week, twice monthly, or monthly dosing for more than two weeks, a month, two months, three months, four months, five months, six months, a year or more).
  • intraveneous administrations e.g., weekly, every other week, twice monthly, or monthly dosing for more than two weeks, a month, two months, three months, four months, five months, six months, a year or more.
  • intrathecal administrations are performed first (e.g., weekly, every other week, twice monthly, monthly, once every two months, once every three months dosing for two weeks, a month, two months, three months, four months, five months, six months, a year or more) followed by intraveneous administrations (e.g., weekly, every other week, twice monthly, or monthly dosing for more than two weeks, a month, two months, three months, four months, five months, six months, a year or more).
  • intraveneous administrations e.g., weekly, every other week, twice monthly, or monthly dosing for more than two weeks, a month, two months, three months, four months, five months, six months, a year or more.
  • the intrathecal administration is used in absence of intravenous administration.
  • the intrathecal administration is used in absence of concurrent immunosuppressive therapy.
  • FIG. 1 illustrates exemplary arylsulfatase A (rhASA) concentration data in serum after IV administration.
  • rhASA arylsulfatase A
  • FIG. 2 illustrates exemplary rhASA concentration data in serum after IT-lumbar administration.
  • FIG. 3 illustrates exemplary rhASA concentration in CSF after IV administration.
  • FIG. 4 illustrates exemplary rhASA concentration in CSF after IT-lumbar administration.
  • FIG. 5 illustrates exemplary analysis of the effect of buffer and pH on the thermal stability of rhASA.
  • FIG. 6 illustrates exemplary SDS-PAGE (Coomassie) analysis of rhASA after two weeks at 40 ⁇ 2° C.
  • FIG. 7 illustrates exemplary SDS-PAGE (Coomassie) analysis of rhASA in IT formulations after 3 months at 5 and 25° C.
  • FIG. 8 depicts exemplary rhASA drug substance and drug product appearance after 48 hours of stirring (Panel A) and shaking (Panel B).
  • FIG. 10 illustrates exemplary data demonstrating the buffering capacity of rhASA drug substance compared to buffer control when titrated with hydrochloric acid.
  • FIG. 11 illustrates exemplary data demonstrating the buffering capacity of rhASA drug substance compared to a buffer control when titrated with 1M sodium hydroxide.
  • FIG. 12 depicts exemplary rhASA samples in saline, pH 6.0 varying by concentration.
  • FIG. 13 illustrates exemplary SEC-HPLC analysis of rhASA (pH 5.5 mobile phase) in 154 mM NaCl, pH 5.9.
  • FIG. 14 illustrates exemplary SEC-HPLC analysis of rhASA (pH 7.0 mobile phase) in 154 mM NaCl, pH 5.9.
  • FIG. 15 illustrates exemplary size exclusion profiles of baseline and 11 month stability samples for rhASA in 154 mM NaCl, pH 5.
  • FIG. 16 depicts exemplary photo-micrographs of brain tissue, meninges, infiltrates (mid and high dose groups, both sexes) after treatment.
  • FIG. 17 depicts exemplary photo-micrographs of brain tissue, meninges, infiltrates (mid and high dose groups, both sexes) after treatment.
  • FIG. 18 depicts exemplary photo-micrographs of brain tissue, perivascular, infiltrates (mid dose males; high dose females) after treatment.
  • FIG. 19 depicts exemplary Alcian blue staining of spinal cord of immunotolerant MLD mice treated with rhASA depicts exemplary results illustrating sulfatide reduction as determined by Alcian blue staining of the cervical spinal cord in animals that received intrathecal injections of recombinant hASA at days 1, 8, 15 and 22 at doses of 520 mg/kg brain weight or vehicle control.
  • treatment with intrathecally injected recombinant hASA resulted in reduction of sulfatide accumulation in the cervical spinal cord.
  • FIG. 20 illustrates exemplary morphometry analysis of Alcian blue stained spinal cord sections from immunotolerant MLD mice treated with rhASA, including exemplary results illustrating optical density of Alcian blue in total spinal cord (T-Spinal Cord), total gray matter (T-GM), lumbar gray matter (L-GM), cervical gray matter (C-GM), total white matter (T-WM), lumbar white matter (L-WM), and cervical white matter (C-WM) as determined by morphometry analysis.
  • T-Spinal Cord total gray matter
  • T-GM total gray matter
  • L-GM lumbar gray matter
  • C-GM cervical gray matter
  • T-WM total white matter
  • C-WM cervical white matter
  • C-WM cervical white matter
  • FIG. 22 illustrates exemplary morphometry Analysis of LAMP staining of brain from immunotolerant MLD mice treated with rhASA depicts exemplary results illustrating LAMP-1 staining intensity in corpus collosum (CC), fimbria (F), cerebellar white matter (CB-WM) and brain stem (BS) of animals treated with 20 mg/kg intravenous rhASA, 300 mg/kg brain weight intrathecal rhASA, 520 mg/kg brain weight intravenous rhASA, or vehicle control.
  • CC corpus collosum
  • F fimbria
  • CB-WM cerebellar white matter
  • BS brain stem
  • FIG. 23 illustrates exemplary concentration of rhASA in brain punches of vehicle-dosed juvenile cynomolgus monkeys following EOW IT dosing for 6-months-main necropsy.
  • FIG. 24 illustrates exemplary concentration of rhASA in brain punches of juvenile cynomolgus monkeys following EOW IT dosing of rhASA at 1.8 mg/dose for 6-months—main necropsy.
  • FIG. 25 illustrates exemplary concentration of rhASA in brain punches of juvenile cynomolgus monkeys following EOW IT Dosing of rhASA at 6.0 mg/dose for 6-months—main necropsy.
  • FIG. 26 illustrates exemplary concentration of rhASA in brain punches of juvenile cynomolgus monkeys following EOW IT dosing of rhASA at 18.6 mg/dose for 6-months—main necropsy.
  • FIG. 27 illustrates exemplary concentration of ASA in brain punches of juvenile cynomolgus monkeys following EOW IT dosing (PBS-control) for 6-months—recovery necropsy.
  • FIG. 28 illustrates exemplary concentration of rhASA in brain punches of juvenile cynomolgus monkeys following EOW IT dosing of vehicle for 6-months—recovery necropsy.
  • FIG. 29 illustrates exemplary concentration of rhASA in brain punches of juvenile cynomolgus monkeys following EOW IT dosing of rhASA at 1.8 mg/dose for 6-months—recovery necropsy
  • FIG. 30 illustrates exemplary concentration of rhASA in brain punches of juvenile cynomolgus monkeys following EOW IT dosing of rhASA at 6.0 mg/dose for 6 months—recovery necropsy
  • FIG. 31 illustrates exemplary concentration of rhASA in brain punches of juvenile cynomolgus following EOW IT dosing of rhASA at 18.6 mg/dose for 6-months—recovery necropsy
  • FIG. 32 illustrates exemplary concentration of rhASA in selected punches from surface of brain for device control, vehicle, 1.8 mg, 6.0 mg and 18.6 mg treated animals. (male and female separate, device control data is from recovery necropsy, all other data from main necropsy).
  • FIG. 33 illustrates exemplary concentration of rhASA in selected punches from deep white area of brain for device control, vehicle, 1.8 mg, 6.0 mg and 18.6 mg treated animals. (male and female separate, device control data is from recovery necropsy, all other data from main necropsy).
  • FIG. 34 illustrates exemplary concentration of rhASA in selected punches from deep grey area of brain for device control, vehicle, 1.8 mg, 6.0 mg and 18.6 mg treated animals. (male and female separate, device control data is from recovery necropsy, all other data from main necropsy).
  • FIG. 35 illustrates exemplary concentration of rhASA in selected punches from various regions in device control, vehicle, 1.8. mg, 6.0 mg and 18.6 mg treated animals. (male and female combined, device control data is from recovery necropsy, all other data from main necropsy).
  • FIG. 36 illustrates exemplary concentration of rhASA in spinal cord sections of juvenile cynomolgus monkeys following EOW IT dosing for 6-months—recovery necroscopy.
  • FIG. 37 illustrates exemplary concentration of rhASA in liver of juvenile cynomolgus monkeys following EOW IT dosing for 6-months—recovery necroscopy.
  • FIG. 38 illustrates exemplary anatomical locations of certain brain punches.
  • FIG. 39 illustrates exemplary anatomical locations of certain brain punches.
  • FIG. 40 illustrates exemplary anatomical locations of certain brain punches.
  • FIG. 41 illustrates exemplary anatomical locations of certain brain punches.
  • FIG. 42 illustrates exemplary anatomical locations of certain brain punches.
  • FIG. 43 illustrates exemplary anatomical locations of certain brain punches.
  • FIG. 44A-G illustrate the concentration of recombinant human arylsulfatase A (rhASA) in extracted tissue punches from the brain tissues of adult and juvenile cynomolgus monkeys administered either a vehicle, 1.8 mg rhASA or 18.6 mg rhASA.
  • rhASA human arylsulfatase A
  • FIGS. 45A and B illustrate exemplary comparison of the concentrations of recombinant human arylsulfatase A (rhASA) detected in the deep white matter ( FIG. 45A ) or in the deep grey matter ( FIG. 45B ) brain tissues of adult and juvenile cynomolgus monkeys which were intrathecally (IT) or intracerebroventricularly (ICV) administered rhASA.
  • rhASA human arylsulfatase A
  • FIG. 46A illustrate concentrations of rhASA detected in several tissue punches obtained from juvenile ( ⁇ 12 months of age) cynomolgus monkeys IT-administered an 18.6 or a 1.8 mg dose of recombinant human arylsulfatase A (rhASA).
  • rhASA recombinant human arylsulfatase A
  • subcortical white matter (1) periventricular white matter and deep white matter (2); subcortical white matter (3); subcortical white matter (4); internal capsule (5); internal capsule caudate nucleus (6); deep white matter (7); subcortical white matter and cortex (8); putamen (9); temporal subcortical white matter and cortex (10), deep grey matter (11), deep grey matter (12), frontal periventricular & subcortical (13); subcortical white matter, cortex superficial perifalxian (14); corpus callosum and pericallosal subcortical white matter (15); deep subcortical white matter (16); deep grey matter (17); deep grey matter (18); periventricular white matter (19); deep subcortical white matter (20); hippocampus (21); corpus callosum (22); deep white matter (23); subcortical white matter, occipital lobe (24); and cerebellar white matter (25).
  • FIG. 47A illustrates the area of deep white matter tissue extracted from a cynomolgus monkey IT-administered 1.8 mg of rhASA.
  • FIG. 47B illustrates immunostaining of the deep white matter tissue and revealed distribution of rhASA in relevant cells.
  • FIG. 47C illustrates that the IT-administered rhASA demonstrated organelle co-localization in the deep white matter tissues of the cynomolgus monkey and in particular in the lysosomes.
  • ASA immunostaining is illustrated in the top left box.
  • FIG. 48 compares the distribution of 124 I-labeled arylsulfatase A (rhASA) using PET scanning 24 hours following either IT- or ICV-administration of such labeled rhASA to a cynomolgus monkey.
  • rhASA I-labeled arylsulfatase A
  • FIG. 49 illustrates the distribution of 124 I-labeled ASA immediately following ICV administration to a cynomolgus monkey, and compares the distribution of IT-administered 124 I-labeled ASA within 2-5 hr.
  • IT administration delivered the 124 I-labeled ASA to the same initial compartments (cisternae and proximal spine) as that shown for the ICV administration.
  • FIG. 50 depicts exemplary ICV and IT administration in a mouse model.
  • FIG. 51 depicts an exemplary intrathecal drug delivery device (IDDD).
  • IDDD intrathecal drug delivery device
  • FIG. 52 depicts an exemplary PORT-A-CATH® low profile intrathecal implantable access system.
  • FIG. 53 depicts an exemplary intrathecal drug delivery device (IDDD).
  • IDDD intrathecal drug delivery device
  • FIG. 54 depicts an exemplary intrathecal drug delivery device (IDDD), which allows for in-home administration for CNS enzyme replacement therapy (ERT).
  • IDDD intrathecal drug delivery device
  • ERT CNS enzyme replacement therapy
  • FIG. 55 illustrates and exemplary diagram of an intrathecal drug delivery device (IDDD) with a securing mechanism.
  • IDDD intrathecal drug delivery device
  • FIG. 56A depicts exemplary locations within a patient's body where an IDDD may be placed;
  • FIG. 56B depicts various components of an intrathecal drug delivery device (IDDD); and
  • FIG. 56C depicts an exemplary insertion location within a patient's body for IT-lumbar injection.
  • IDDD intrathecal drug delivery device
  • Amelioration is meant the prevention, reduction or palliation of a state, or improvement of the state of a subject. Amelioration includes, but does not require complete recovery or complete prevention of a disease condition. In some embodiments, amelioration includes increasing levels of relevant protein or its activity that is deficient in relevant disease tissues.
  • biologically active refers to a characteristic of any agent that has activity in a biological system, and particularly in an organism. For instance, an agent that, when administered to an organism, has a biological effect on that organism, is considered to be biologically active.
  • an agent that, when administered to an organism, has a biological effect on that organism is considered to be biologically active.
  • a portion of that protein or polypeptide that shares at least one biological activity of the protein or polypeptide is typically referred to as a “biologically active” portion.
  • Bulking agent refers to a compound which adds mass to the lyophilized mixture and contributes to the physical structure of the lyophilized cake (e.g., facilitates the production of an essentially uniform lyophilized cake which maintains an open pore structure).
  • exemplary bulking agents include mannitol, glycine, sodium chloride, hydroxyethyl starch, lactose, sucrose, trehalose, polyethylene glycol and dextran.
  • Cation-independent mannose-6-phosphate receptor As used herein, the term “cation-independent mannose-6-phosphate receptor (CI-MPR)” refers to a cellular receptor that binds mannose-6-phosphate (M6P) tags on acid hydrolase precursors in the Golgi apparatus that are destined for transport to the lysosome. In addition to mannose-6-phosphates, the CI-MPR also binds other proteins including IGF-II.
  • the CI-MPR is also known as “M6P/IGF-II receptor,” “CI-MPR/IGF-II receptor,” “IGF-II receptor” or “IGF2 Receptor.” These terms and abbreviations thereof are used interchangeably herein.
  • Concurrent immunosuppressant therapy includes any immunosuppressant therapy used as pre-treatment, preconditioning or in parallel to a treatment method.
  • Diluent refers to a pharmaceutically acceptable (e.g., safe and non-toxic for administration to a human) diluting substance useful for the preparation of a reconstituted formulation.
  • exemplary diluents include sterile water, bacteriostatic water for injection (BWFI), a pH buffered solution (e.g. phosphate-buffered saline), sterile saline solution, Ringer's solution or dextrose solution.
  • Dosage form As used herein, the terms “dosage form” and “unit dosage form” refer to a physically discrete unit of a therapeutic protein for the patient to be treated. Each unit contains a predetermined quantity of active material calculated to produce the desired therapeutic effect. It will be understood, however, that the total dosage of the composition will be decided by the attending physician within the scope of sound medical judgment.
  • Enzyme replacement therapy refers to any therapeutic strategy that corrects an enzyme deficiency by providing the missing enzyme.
  • the missing enzyme is provided by intrathecal administration.
  • the missing enzyme is provided by infusing into bloodsteam. Once administered, enzyme is taken up by cells and transported to the lysosome, where the enzyme acts to eliminate material that has accumulated in the lysosomes due to the enzyme deficiency.
  • the therapeutic enzyme is delivered to lysosomes in the appropriate cells in target tissues where the storage defect is manifest.
  • the terms “improve,” “increase” or “reduce,” or grammatical equivalents indicate values that are relative to a baseline measurement, such as a measurement in the same individual prior to initiation of the treatment described herein, or a measurement in a control individual (or multiple control individuals) in the absence of the treatment described herein.
  • a “control individual” is an individual afflicted with the same form of lysosomal storage disease as the individual being treated, who is about the same age as the individual being treated (to ensure that the stages of the disease in the treated individual and the control individual(s) are comparable).
  • the terms “subject,” “individual” or “patient” refer to a human or a non-human mammalian subject.
  • the individual (also referred to as “patient” or “subject”) being treated is an individual (fetus, infant, child, adolescent, or adult human) suffering from a disease.
  • Intrathecal administration refers to an injection into the spinal canal (intrathecal space surrounding the spinal cord). Various techniques may be used including, without limitation, lateral cerebroventricular injection through a burrhole or cisternal or lumbar puncture or the like.
  • “intrathecal administration” or “intrathecal delivery” according to the present invention refers to IT administration or delivery via the lumbar area or region, i.e., lumbar IT administration or delivery.
  • lumbar region or “lumbar area” refers to the area between the third and fourth lumbar (lower back) vertebrae and, more inclusively, the L2-S1 region of the spine.
  • Linker refers to, in a fusion protein, an amino acid sequence other than that appearing at a particular position in the natural protein and is generally designed to be flexible or to interpose a structure, such as an a-helix, between two protein moieties.
  • a linker is also referred to as a spacer.
  • Lyoprotectant refers to a molecule that prevents or reduces chemical and/or physical instability of a protein or other substance upon lyophilization and subsequent storage.
  • exemplary lyoprotectants include sugars such as sucrose or trehalose; an amino acid such as monosodium glutamate or histidine; a methylamine such as betaine; a lyotropic salt such as magnesium sulfate: a polyol such as trihydric or higher sugar alcohols, e.g.
  • a lyoprotectant is a non-reducing sugar, such as trehalose or sucrose.
  • Lysosomal enzyme refers to any enzyme that is capable of reducing accumulated materials in mammalian lysosomes or that can rescue or ameliorate one or more lysosomal storage disease symptoms.
  • Lysosomal enzymes suitable for the invention include both wild-type or modified lysosomal enzymes and can be produced using recombinant and synthetic methods or purified from nature sources. Exemplary lysosomal enzymes are listed in Table 1.
  • Lysosomal enzyme deficiency refers to a group of genetic disorders that result from deficiency in at least one of the enzymes that are required to break macromolecules (e.g., enzyme substartes) down to peptides, amino acids, monosaccharides, nucleic acids and fatty acids in lysosomes.
  • macromolecules e.g., enzyme substartes
  • peptides amino acids, monosaccharides, nucleic acids and fatty acids in lysosomes.
  • individuals suffering from lysosomal enzyme deficiencies have accumulated materials in various tissues (e.g., CNS, liver, spleen, gut, blood vessel walls and other organs).
  • Lysosomal storage disease refers to any disease resulting from the deficiency of one or more lysosomal enzymes necessary for metabolizing natural macromolecules. These diseases typically result in the accumulation of un-degraded molecules in the lysosomes, resulting in increased numbers of storage granules (also termed storage vesicles). These diseases and various examples are described in more detail below.
  • Polypeptide As used herein, a “polypeptide”, generally speaking, is a string of at least two amino acids attached to one another by a peptide bond. In some embodiments, a polypeptide may include at least 3-5 amino acids, each of which is attached to others by way of at least one peptide bond. Those of ordinary skill in the art will appreciate that polypeptides sometimes include “non-natural” amino acids or other entities that nonetheless are capable of integrating into a polypeptide chain, optionally.
  • replacement enzyme refers to any enzyme that can act to replace at least in part the deficient or missing enzyme in a disease to be treated.
  • replacement enzyme refers to any enzyme that can act to replace at least in part the deficient or missing lysosomal enzyme in a lysosomal storage disease to be treated.
  • a replacement enzyme is capable of reducing accumulated materials in mammalian lysosomes or that can rescue or ameliorate one or more lysosomal storage disease symptoms.
  • Replacement enzymes suitable for the invention include both wild-type or modified lysosomal enzymes and can be produced using recombinant and synthetic methods or purified from nature sources.
  • a replacement enzyme can be a recombinant, synthetic, gene-activated or natural enzyme.
  • Soluble refers to the ability of a therapeutic agent to form a homogenous solution.
  • the solubility of the therapeutic agent in the solution into which it is administered and by which it is transported to the target site of action is sufficient to permit the delivery of a therapeutically effective amount of the therapeutic agent to the targeted site of action.
  • target site of action e.g., the cells and tissues of the brain
  • solubility of the therapeutic agents include ionic strength, amino acid sequence and the presence of other co-solubilizing agents or salts (e.g., calcium salts).
  • the pharmaceutical compositions are formulated such that calcium salts are excluded from such compositions.
  • therapeutic agents in accordance with the present invention are soluble in its corresponding pharmaceutical composition.
  • isotonic solutions are generally preferred for parenterally administered drugs, the use of isotonic solutions may limit adequate solubility for some therapeutic agents and, in particular some proteins and/or enzymes.
  • Slightly hypertonic solutions e.g., up to 175 mM sodium chloride in 5 mM sodium phosphate at pH 7.0
  • sugar-containing solutions e.g., up to 2% sucrose in 5 mM sodium phosphate at pH 7.0
  • the most common approved CNS bolus formulation composition is saline (150 mM NaCl in water).
  • Stability refers to the ability of the therapeutic agent (e.g., a recombinant enzyme) to maintain its therapeutic efficacy (e.g., all or the majority of its intended biological activity and/or physiochemical integrity) over extended periods of time.
  • the stability of a therapeutic agent, and the capability of the pharmaceutical composition to maintain stability of such therapeutic agent may be assessed over extended periods of time (e.g., for at least 1, 3, 6, 12, 18, 24, 30, 36 months or more).
  • pharmaceutical compositions described herein have been formulated such that they are capable of stabilizing, or alternatively slowing or preventing the degradation, of one or more therapeutic agents formulated therewith (e.g., recombinant proteins).
  • a stable formulation is one in which the therapeutic agent therein essentially retains its physical and/or chemical integrity and biological activity upon storage and during processes (such as freeze/thaw, mechanical mixing and lyophilization).
  • HMW high molecular weight
  • subject means any mammal, including humans. In certain embodiments of the present invention the subject is an adult, an adolescent or an infant. Also contemplated by the present invention are the administration of the pharmaceutical compositions and/or performance of the methods of treatment in-utero.
  • Substantial homology is used herein to refer to a comparison between amino acid or nucleic acid sequences. As will be appreciated by those of ordinary skill in the art, two sequences are generally considered to be “substantially homologous” if they contain homologous residues in corresponding positions. Homologous residues may be identical residues. Alternatively, homologous residues may be non-identical residues will appropriately similar structural and/or functional characteristics.
  • amino acids are typically classified as “hydrophobic” or “hydrophilic” amino acids, and/or as having “polar” or “non-polar” side chains Substitution of one amino acid for another of the same type may often be considered a “homologous” substitution.
  • amino acid or nucleic acid sequences may be compared using any of a variety of algorithms, including those available in commercial computer programs such as BLASTN for nucleotide sequences and BLASTP, gapped BLAST, and PSI-BLAST for amino acid sequences.
  • Exemplary such programs are described in Altschul, et al., Basic local alignment search tool, J. Mol. Biol., 215(3): 403-410, 1990; Altschul, et al., Methods in Enzymology ; Altschul, et al., “Gapped BLAST and PSI-BLAST: a new generation of protein database search programs”, Nucleic Acids Res.
  • two sequences are considered to be substantially homologous if at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more of their corresponding residues are homologous over a relevant stretch of residues.
  • the relevant stretch is a complete sequence.
  • the relevant stretch is at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500 or more residues.
  • amino acid or nucleic acid sequences may be compared using any of a variety of algorithms, including those available in commercial computer programs such as BLASTN for nucleotide sequences and BLASTP, gapped BLAST, and PSI-BLAST for amino acid sequences. Exemplary such programs are described in Altschul, et al., Basic local alignment search tool, J. Mol.
  • two sequences are considered to be substantially identical if at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more of their corresponding residues are identical over a relevant stretch of residues.
  • the relevant stretch is a complete sequence.
  • the relevant stretch is at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500 or more residues.
  • Synthetic CSF refers to a solution that has pH, electrolyte composition, glucose content and osmalarity consistent with the cerebrospinal fluid. Synthetic CSF is also referred to as artificial CSF. In some embodiments, synthetic CSF is an Elliott's B solution.
  • Suitable for CNS delivery As used herein, the phrase “suitable for CNS delivery” or “suitable for intrathecal delivery” as it relates to the pharmaceutical compositions of the present invention generally refers to the stability, tolerability, and solubility properties of such compositions, as well as the ability of such compositions to deliver an effective amount of the therapeutic agent contained therein to the targeted site of delivery (e.g., the CSF or the brain).
  • Target tissues refers to any tissue that is affected by the lysosomal storage disease to be treated or any tissue in which the deficient lysosomal enzyme is normally expressed.
  • target tissues include those tissues in which there is a detectable or abnormally high amount of enzyme substrate, for example stored in the cellular lysosomes of the tissue, in patients suffering from or susceptible to the lysosomal storage disease.
  • target tissues include those tissues that display disease-associated pathology, symptom, or feature.
  • target tissues include those tissues in which the deficient lysosomal enzyme is normally expressed at an elevated level.
  • a target tissue may be a brain target tissue, a spinal cord target tissue and/or a peripheral target tissue. Exemplary target tissues are described in detail below.
  • therapeutic moiety refers to a portion of a molecule that renders the therapeutic effect of the molecule.
  • a therapeutic moiety is a polypeptide having therapeutic activity.
  • therapeutically effective amount refers to an amount of a therapeutic protein (e.g., replacement enzyme) which confers a therapeutic effect on the treated subject, at a reasonable benefit/risk ratio applicable to any medical treatment.
  • the therapeutic effect may be objective (i.e., measurable by some test or marker) or subjective (i.e., subject gives an indication of or feels an effect).
  • therapeutically effective amount refers to an amount of a therapeutic protein or composition effective to treat, ameliorate, or prevent a desired disease or condition, or to exhibit a detectable therapeutic or preventative effect, such as by ameliorating symptoms associated with the disease, preventing or delaying the onset of the disease, and/or also lessening the severity or frequency of symptoms of the disease.
  • a therapeutically effective amount is commonly administered in a dosing regimen that may comprise multiple unit doses.
  • a therapeutically effective amount (and/or an appropriate unit dose within an effective dosing regimen) may vary, for example, depending on route of administration, on combination with other pharmaceutical agents.
  • the specific therapeutically effective amount (and/or unit dose) for any particular patient may depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific pharmaceutical agent employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and/or rate of excretion or metabolism of the specific fusion protein employed; the duration of the treatment; and like factors as is well known in the medical arts.
  • Tolerable refers to the ability of the pharmaceutical compositions of the present invention to not elicit an adverse reaction in the subject to whom such composition is administered, or alternatively not to elicit a serious adverse reaction in the subject to whom such composition is administered. In some embodiments, the pharmaceutical compositions of the present invention are well tolerated by the subject to whom such compositions is administered.
  • treatment refers to any administration of a therapeutic protein (e.g., lysosomal enzyme) that partially or completely alleviates, ameliorates, relieves, inhibits, delays onset of, reduces severity of and/or reduces incidence of one or more symptoms or features of a particular disease, disorder, and/or condition (e.g., Hunters syndrome, Sanfilippo B syndrome).
  • a therapeutic protein e.g., lysosomal enzyme
  • Such treatment may be of a subject who does not exhibit signs of the relevant disease, disorder and/or condition and/or of a subject who exhibits only early signs of the disease, disorder, and/or condition.
  • Such treatment may be of a subject who exhibits one or more established signs of the relevant disease, disorder and/or condition.
  • the present invention provides, among other things, improved methods and compositions for effective direct delivery of a therapeutic agent to the central nervous system (CNS).
  • a replacement enzyme e.g., an ASA protein
  • a lysososmal storage disease e.g., Metachromatic Leukodystrophy Disease
  • CS F cerebrospinal fluid
  • the present inventors found that the replacement enzyme may be delivered in a simple saline or buffer-based formulation, without using synthetic CSF.
  • intrathecal delivery according to the present invention does not result in substantial adverse effects, such as severe immune response, in the subject. Therefore, in some embodiments, intrathecal delivery according to the present invention may be used in absence of concurrent immunosuppressant therapy (e.g., without induction of immune tolerance by pre-treatment or pre-conditioning).
  • intrathecal delivery according to the present invention permits efficient diffusion across various brain tissues resulting in effective delivery of the replacement enzyme in various target brain tissues in surface, shallow and/or deep brain regions. In some embodiments, intrathecal delivery according to the present invention resulted in sufficient amount of replacement enzymes entering the peripheral circulation. As a result, in some cases, intrathecal delivery according to the present invention resulted in delivery of the replacement enzyme in peripheral tissues, such as liver, heart, spleen and kidney. This discovery is unexpected and can be particular useful for the treatment of lysosomal storage diseases that have both CNS and peripheral components, which would typically require both regular intrathecal administration and intravenous administration. It is contemplated that intrathecal delivery according to the present invention may allow reduced dosing and/or frequency of iv injection without compromising therapeutic effects in treating peripheral symptoms.
  • the present invention provides various unexpected and beneficial features that allow efficient and convenient delivery of replacement enzymes to various brain target tissues, resulting in effective treatment of lysosomal storage diseases that have CNS indications.
  • inventive methods and compositions provided by the present invention are used to deliver an arylsulfatase A (ASA) protein to the CNS for treatment of Metachromatic Leukodystrophy Disease.
  • a suitable ASA protein can be any molecule or a portion of a molecule that can substitute for naturally-occurring arylsulfatase A (ASA) protein activity or rescue one or more phenotypes or symptoms associated with ASA-deficiency.
  • a replacement enzyme suitable for the invention is a polypeptide having an N-terminus and a C-terminus and an amino acid sequence substantially similar or identical to mature human ASA protein.
  • human ASA is produced as a precursor molecule that is processed to a mature form. This process generally occurs by removing the 18 amino acid signal peptide.
  • the precursor form is also referred to as full-length precursor or full-length ASA protein, which contains 507 amino acids.
  • the N-terminal 18 amino acids are cleaved, resulting in a mature form that is 489 amino acids in length. Thus, it is contemplated that the N-terminal 18 amino acids is generally not required for the ASA protein activity.
  • the amino acid sequences of the mature form (SEQ ID NO:1) and full-length precursor (SEQ ID NO:2) of a typical wild-type or naturally-occurring human ASA protein are shown in Table 1.
  • a therapeutic moiety suitable for the present invention is mature human ASA protein (SEQ ID NO:1).
  • a suitable therapeutic moiety may be a homologue or an analogue of mature human ASA protein.
  • a homologue or an analogue of mature human ASA protein may be a modified mature human ASA protein containing one or more amino acid substitutions, deletions, and/or insertions as compared to a wild-type or naturally-occurring ASA protein (e.g., SEQ ID NO:1), while retaining substantial ASA protein activity.
  • a therapeutic moiety suitable for the present invention is substantially homologous to mature human ASA protein (SEQ ID NO:1).
  • a therapeutic moiety suitable for the present invention has an amino acid sequence at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%. 93%, 94%, 95%, 96%, 97%, 98%, 99% or more homologous to SEQ ID NO:1.
  • a therapeutic moiety suitable for the present invention is substantially identical to mature human ASA protein (SEQ ID NO:1).
  • a therapeutic moiety suitable for the present invention has an amino acid sequence at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to SEQ ID NO:1.
  • a therapeutic moiety suitable for the present invention contains a fragment or a portion of mature human ASA protein.
  • a replacement enzyme suitable for the present invention is full-length ASA protein.
  • a suitable replacement enzyme may be a homologue or an analogue of full-length human ASA protein.
  • a homologue or an analogue of full-length human ASA protein may be a modified full-length human ASA protein containing one or more amino acid substitutions, deletions, and/or insertions as compared to a wild-type or naturally-occurring full-length ASA protein (e.g., SEQ ID NO:2), while retaining substantial ASA protein activity.
  • a replacement enzyme suitable for the present invention is substantially homologous to full-length human ASA protein (SEQ ID NO:2).
  • a replacement enzyme suitable for the present invention has an amino acid sequence at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more homologous to SEQ ID NO:2.
  • a replacement enzyme suitable for the present invention is substantially identical to SEQ ID NO:2.
  • a replacement enzyme suitable for the present invention has an amino acid sequence at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to SEQ ID NO:2.
  • a replacement enzyme suitable for the present invention contains a fragment or a portion of full-length human ASA protein. As used herein, a full-length ASA protein typically contains signal peptide sequence.
  • a therapeutic protein includes a targeting moiety (e.g., a lysosome targeting sequence) and/or a membrane-penetrating peptide.
  • a targeting sequence and/or a membrane-penetrating peptide is an intrinsic part of the therapeutic moiety (e.g., via a chemical linkage, via a fusion protein).
  • a targeting sequence contains a mannose-6-phosphate moiety.
  • a targeting sequence contains an IGF-I moiety.
  • a targeting sequence contains an IGF-II moiety.
  • inventive methods and compositions according to the present invention can be used to treat other lysosomal storage diseases, in particular those lysosomal storage diseases having CNS etiology and/or symptoms, including, but are not limited to, aspartylglucosaminuria, cholesterol ester storage disease, Wolman disease, cystinosis, Danon disease, Fabry disease, Farber lipogranulomatosis, Farber disease, fucosidosis, galactosialidosis types I/II, Gaucher disease types I/II/III, globoid cell leukodystrophy, Krabbe disease, glycogen storage disease II, Pompe disease, GM1-gangliosidosis types I/II/III, GM2-gangliosidosis type I, Tay Sachs disease, GM2-gangliosidosis type II, Sandhoff disease, GM2-gangliosidosis, ⁇ -mannosidosis types I/II, .beta.-mannos
  • replacement enzymes suitable for the present invention may include any enzyme that can act to replace at least partial activity of the deficient or missing lysosomal enzyme in a lysosomal storage disease to be treated.
  • a replacement enzyme is capable of reducing accumulated substance in lysosomes or that can rescue or ameliorate one or more lysosomal storage disease symptoms.
  • a suitable replacement enzyme may be any lysosomal enzyme known to be associated with the lysosomal storage disease to be treated.
  • a suitable replacement enzyme is an enzyme selected from the enzyme listed in Table 2 above.
  • a replacement enzyme suitable for the invention may have a wild-type or naturally occurring sequence.
  • a replacement enzyme suitable for the invention may have a modified sequence having substantial homology or identify to the wild-type or naturally-occurring sequence (e.g., having at least 50%, 55%, 60%, 65%, 70%. 75%, 80%, 85%, 90%, 95%, 98% sequence identity to the wild-type or naturally-occurring sequence).
  • a replacement enzyme suitable for the present invention may be produced by any available means.
  • replacement enzymes may be recombinantly produced by utilizing a host cell system engineered to express a replacement enzyme-encoding nucleic acid.
  • replacement enzymes may be produced by activating endogenous genes.
  • replacement enzymes may be partially or fully prepared by chemical synthesis.
  • replacements enzymes may also be purified from natural sources.
  • any expression system can be used.
  • known expression systems include, for example, egg, baculovirus, plant, yeast, or mammalian cells.
  • enzymes suitable for the present invention are produced in mammalian cells.
  • mammalian cells that may be used in accordance with the present invention include BALB/c mouse myeloma line (NSO/1, ECACC No: 85110503); human retinoblasts (PER.C6, CruCell, Leiden, The Netherlands); monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J.
  • human fibrosarcoma cell line e.g., HT1080
  • baby hamster kidney cells BHK, ATCC CCL 10
  • Chinese hamster ovary cells +/ ⁇ DHFR CHO, Urlaub and Chasin, Proc. Natl. Acad. Sci. USA, 77:4216, 1980
  • mouse sertoli cells TM4, Mather, Biol.
  • monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1 587); human cervical carcinoma cells (HeLa, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al., Annals N.Y. Acad. Sci., 383:44-68, 1982); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2).
  • inventive methods according to the present invention are used to deliver replacement enzymes produced from human cells. In some embodiments, inventive methods according to the present invention are used to deliver replacement enzymes produced from CHO cells.
  • replacement enzymes delivered using a method of the invention contain a moiety that binds to a receptor on the surface of brain cells to facilitate cellular uptake and/or lysosomal targeting.
  • a receptor may be the cation-independent mannose-6-phosphate receptor (CI-MPR) which binds the mannose-6-phosphate (M6P) residues.
  • the CI-MPR also binds other proteins including IGF-II.
  • a replacement enzyme suitable for the present invention contains M6P residues on the surface of the protein.
  • a replacement enzyme suitable for the present invention may contain bis-phosphorylated oligosaccharides which have higher binding affinity to the CI-MPR.
  • a suitable enzyme contains up to about an average of about at least 20% bis-phosphorylated oligosaccharides per enzyme. In other embodiments, a suitable enzyme may contain about 10%, 15%, 18%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60% bis-phosphorylated oligosaccharides per enzyme. While such bis-phosphorylated oligosaccharides may be naturally present on the enzyme, it should be noted that the enzymes may be modified to possess such oligosaccharides.
  • suitable replacement enzymes may be modified by certain enzymes which are capable of catalyzing the transfer of N-acetylglucosamine-L-phosphate from UDP-GlcNAc to the 6′ position of ⁇ -1,2-linked mannoses on lysosomal enzymes.
  • Methods and compositions for producing and using such enzymes are described by, for example, Canfield et al. in U.S. Pat. No. 6,537,785, and U.S. Pat. No. 6,534,300, each incorporated herein by reference.
  • replacement enzymes for use in the present invention may be conjugated or fused to a lysosomal targeting moiety that is capable of binding to a receptor on the surface of brain cells.
  • a suitable lysosomal targeting moiety can be IGF-I, RAP, p97, and variants, homologues or fragments thereof (e.g., including those peptide having a sequence at least 70%, 75%, 80%, 85%, 90%, or 95% identical to a wild-type mature human IGF-I, IGF-II, RAP, p97 peptide sequence).
  • replacement enzymes suitable for the present invention have not been modified to enhance delivery or transport of such agents across the BBB and into the CNS.
  • a therapeutic protein includes a targeting moiety (e.g., a lysosome targeting sequence) and/or a membrane-penetrating peptide.
  • a targeting sequence and/or a membrane-penetrating peptide is an intrinsic part of the therapeutic moiety (e.g., via a chemical linkage, via a fusion protein).
  • a targeting sequence contains a mannose-6-phosphate moiety.
  • a targeting sequence contains an IGF-I moiety.
  • a targeting sequence contains an IGF-II moiety.
  • Aqueous pharmaceutical solutions and compositions that are traditionally used to deliver therapeutic agents to the CNS of a subject include unbuffered isotonic saline and Elliott's B solution, which is artificial CSF.
  • a comparison depicting the compositions of CSF relative to Elliott's B solution is included in Table 3 below.
  • the concentration of Elliot's B Solution closely parallels that of the CSF.
  • Elliott's B Solution however contains a very low buffer concentration and accordingly may not provide the adequate buffering capacity needed to stabilize therapeutic agents (e.g., proteins), especially over extended periods of time (e.g., during storage conditions).
  • Directional B Solution contains certain salts which may be incompatible with the formulations intended to deliver some therapeutic agents, and in particular proteins or enzymes.
  • the calcium salts present in Elliott's B Solution are capable of mediating protein precipitation and thereby reducing the stability of the formulation.
  • the present invention provides formulations, in either aqueous, pre-lyophilized, lyophilized or reconstituted form, for therapeutic agents that have been formulated such that they are capable of stabilizing, or alternatively slowing or preventing the degradation, of one or more therapeutic agents formulated therewith (e.g., recombinant proteins).
  • the present formulations provide lyophilization formulation for therapeutic agents.
  • the present formulations provide aqueous formulations for therapeutic agents.
  • the formulations are stable formulations.
  • stable refers to the ability of the therapeutic agent (e.g., a recombinant enzyme) to maintain its therapeutic efficacy (e.g., all or the majority of its intended biological activity and/or physiochemical integrity) over extended periods of time.
  • the stability of a therapeutic agent, and the capability of the pharmaceutical composition to maintain stability of such therapeutic agent may be assessed over extended periods of time (e.g., preferably for at least 1, 3, 6, 12, 18, 24, 30, 36 months or more).
  • a stable formulation is one in which the therapeutic agent therein essentially retains its physical and/or chemical integrity and biological activity upon storage and during processes (such as freeze/thaw, mechanical mixing and lyophilization).
  • HMW high molecular weight
  • Stability of the therapeutic agent is of particular importance with respect to the maintenance of the specified range of the therapeutic agent concentration required to enable the agent to serve its intended therapeutic function. Stability of the therapeutic agent may be further assessed relative to the biological activity or physiochemical integrity of the therapeutic agent over extended periods of time. For example, stability at a given time point may be compared against stability at an earlier time point (e.g., upon formulation day 0) or against unformulated therapeutic agent and the results of this comparison expressed as a percentage.
  • the pharmaceutical compositions of the present invention maintain at least 100%, at least 99%, at least 98%, at least 97% at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55% or at least 50% of the therapeutic agent's biological activity or physiochemical integrity over an extended period of time (e.g., as measured over at least about 6-12 months, at room temperature or under accelerated storage conditions).
  • the therapeutic agents are preferably soluble in the pharmaceutical compositions of the present invention.
  • the term “soluble” as it relates to the therapeutic agents of the present invention refer to the ability of such therapeutic agents to form a homogenous solution.
  • the solubility of the therapeutic agent in the solution into which it is administered and by which it is transported to the target site of action is sufficient to permit the delivery of a therapeutically effective amount of the therapeutic agent to the targeted site of action.
  • relevant factors which may impact protein solubility include ionic strength, amino acid sequence and the presence of other co-solubilizing agents or salts (e.g., calcium salts.)
  • the pharmaceutical compositions are formulated such that calcium salts are excluded from such compositions.
  • Suitable formulations in either aqueous, pre-lyophilized, lyophilized or reconstituted form, may contain a therapeutic agent of interest at various concentrations.
  • formulations may contain a protein or therapeutic agent of interest at a concentration in the range of about 0.1 mg/ml to 100 mg/ml (e.g., about 0.1 mg/ml to 80 mg/ml, about 0.1 mg/ml to 60 mg/ml, about 0.1 mg/ml to 50 mg/ml, about 0.1 mg/ml to 40 mg/ml, about 0.1 mg/ml to 30 mg/ml, about 0.1 mg/ml to 25 mg/ml, about 0.1 mg/ml to 20 mg/ml, about 0.1 mg/ml to 60 mg/ml, about 0.1 mg/ml to 50 mg/ml, about 0.1 mg/ml to 40 mg/ml, about 0.1 mg/ml to 30 mg/ml, about 0.1 mg/ml to 25 mg/ml, about 0.1 mg/
  • formulations according to the invention may contain a therapeutic agent at a concentration of approximately 1 mg/ml, 5 mg/ml, 10 mg/ml, 15 mg/ml, 20 mg/ml, 25 mg/ml, 30 mg/ml, 40 mg/ml, 50 mg/ml, 60 mg/ml, 70 mg/ml, 80 mg/ml, 90 mg/ml, or 100 mg/ml.
  • the formulations of the present invention are characterized by their tolerability either as aqueous solutions or as reconstituted lyophilized solutions.
  • the terms “tolerable” and “tolerability” refer to the ability of the pharmaceutical compositions of the present invention to not elicit an adverse reaction in the subject to whom such composition is administered, or alternatively not to elicit a serious adverse reaction in the subject to whom such composition is administered.
  • the pharmaceutical compositions of the present invention are well tolerated by the subject to whom such compositions is administered.
  • the pH of the formulation is an additional factor which is capable of altering the solubility of a therapeutic agent (e.g., an enzyme or protein) in an aqueous formulation or for a pre-lyophilization formulation.
  • a therapeutic agent e.g., an enzyme or protein
  • the formulations of the present invention preferably comprise one or more buffers.
  • the aqueous formulations comprise an amount of buffer sufficient to maintain the optimal pH of said composition between about 4.0-8.0 (e.g., about 4.0, 4.5, 5.0, 5.5, 6.0, 6.2, 6.4, 6.5, 6.6, 6.8, 7.0, 7.5, or 8.0).
  • the pH of the formulation is between about 5.0-7.5, between about 5.5-7.0, between about 6.0-7.0, between about 5.5-6.0, between about 5.5-6.5, between about 5.0-6.0, between about 5.0-6.5 and between about 6.0-7.5.
  • Suitable buffers include, for example acetate, citrate, histidine, phosphate, succinate, tris(hydroxymethyl)aminomethane (“Tris”) and other organic acids.
  • Tris tris(hydroxymethyl)aminomethane
  • a buffering agent is present at a concentration ranging between about 1 mM to about 150 mM, or between about 10 mM to about 50 mM, or between about 15 mM to about 50 mM, or between about 20 mM to about 50 mM, or between about 25 mM to about 50 mM.
  • a suitable buffering agent is present at a concentration of approximately 1 mM, 5 mM, 10 mM, 15 mM, 20 mM, 25 mM, 30 mM, 35 mM, 40 mM, 45 mM 50 mM, 75 mM, 100 mM, 125 mM or 150 mM.
  • formulations in either aqueous, pre-lyophilized, lyophilized or reconstituted form, contain an isotonicity agent to keep the formulations isotonic.
  • isotonic is meant that the formulation of interest has essentially the same osmotic pressure as human blood.
  • Isotonic formulations will generally have an osmotic pressure from about 240 mOsm/kg to about 350 mOsm/kg. Isotonicity can be measured using, for example, a vapor pressure or freezing point type osmometers.
  • Exemplary isotonicity agents include, but are not limited to, glycine, sorbitol, mannitol, sodium chloride and arginine.
  • suitable isotonic agents may be present in aqueous and/or pre-lyophilized formulations at a concentration from about 0.01-5% (e.g., 0.05, 0.1, 0.15, 0.2, 0.3, 0.4, 0.5, 0.75, 1.0, 1.25, 1.5, 2.0, 2.5, 3.0, 4.0 or 5.0%) by weight.
  • formulations for lyophilization contain an isotonicity agent to keep the pre-lyophilization formulations or the reconstituted formulations isotonic.
  • isotonic solutions While generally isotonic solutions are preferred for parenterally administered drugs, the use of isotonic solutions may change solubility for some therapeutic agents and in particular some proteins and/or enzymes. Slightly hypertonic solutions (e.g., up to 175 mM sodium chloride in 5 mM sodium phosphate at pH 7.0) and sugar-containing solutions (e.g., up to 2% sucrose in 5 mM sodium phosphate at pH 7.0) have been demonstrated to be well tolerated. The most common approved CNS bolus formulation composition is saline (about 150 mM NaCl in water).
  • formulations may contain a stabilizing agent, or lyoprotectant, to protect the protein.
  • a suitable stabilizing agent is a sugar, a non-reducing sugar and/or an amino acid.
  • sugars include, but are not limited to, dextran, lactose, mannitol, mannose, sorbitol, raffinose, sucrose and trehalose.
  • exemplary amino acids include, but are not limited to, arginine, glycine and methionine.
  • Additional stabilizing agents may include sodium chloride, hydroxyethyl starch and polyvinylpyrolidone. The amount of stabilizing agent in the lyophilized formulation is generally such that the formulation will be isotonic.
  • hypertonic reconstituted formulations may also be suitable.
  • the amount of stabilizing agent must not be too low such that an unacceptable amount of degradation/aggregation of the therapeutic agent occurs.
  • Exemplary stabilizing agent concentrations in the formulation may range from about 1 mM to about 400 mM (e.g., from about 30 mM to about 300 mM, and from about 50 mM to about 100 mM), or alternatively, from 0.1% to 15% (e.g., from 1% to 10%, from 5% to 15%, from 5% to 10%) by weight.
  • the ratio of the mass amount of the stabilizing agent and the therapeutic agent is about 1:1.
  • the ratio of the mass amount of the stabilizing agent and the therapeutic agent can be about 0.1:1, 0.2:1, 0.25:1, 0.4:1, 0.5:1, 1:1, 2:1, 2.6:1, 3:1, 4:1, 5:1, 10:1, or 20:1.
  • the stabilizing agent is also a lyoprotectant.
  • liquid formulations suitable for the present invention contain amorphous materials. In some embodiments, liquid formulations suitable for the present invention contain a substantial amount of amorphous materials (e.g., sucrose-based formulations). In some embodiments, liquid formulations suitable for the present invention contain partly crystalline/partly amorphous materials.
  • suitable formulations for lyophilization may further include one or more bulking agents.
  • a “bulking agent” is a compound which adds mass to the lyophilized mixture and contributes to the physical structure of the lyophilized cake.
  • a bulking agent may improve the appearance of lyophilized cake (e.g., essentially uniform lyophilized cake).
  • Suitable bulking agents include, but are not limited to, sodium chloride, lactose, mannitol, glycine, sucrose, trehalose, hydroxyethyl starch.
  • concentrations of bulking agents are from about 1% to about 10% (e.g., 1.0%, 1.5%, 2.0%, 2.5%, 3.0%, 3.5%, 4.0%, 4.5%, 5.0%, 5.5%, 6.0%, 6.5%, 7.0%, 7.5%, 8.0%, 8.5%, 9.0%, 9.5%, and 10.0%).
  • a surfactant include nonionic surfactants such as Polysorbates (e.g., Polysorbates 20 or 80); poloxamers (e.g., poloxamer 188); Triton; sodium dodecyl sulfate (SDS); sodium laurel sulfate; sodium octyl glycoside; lauryl-, myristyl-, linoleyl-, or stearyl-sulfobetaine; lauryl-, myristyl-, linoleyl- or stearyl-sarcosine; linoleyl-, myristyl-, or cetyl-betaine; lauroamidopropyl-, cocamidopropyl-, linoleamidopropyl-, myristamidopropyl-, palmidopropyl-, or isostearamidopropyl
  • nonionic surfactants such as Polysorbates (e.g.
  • the amount of surfactant added is such that it reduces aggregation of the protein and minimizes the formation of particulates or effervescences.
  • a surfactant may be present in a formulation at a concentration from about 0.001-0.5% (e.g., about 0.005-0.05%, or 0.005-0.01%).
  • a surfactant may be present in a formulation at a concentration of approximately 0.005%, 0.01%, 0.02%, 0.1%, 0.2%, 0.3%, 0.4%, or 0.5%, etc.
  • the surfactant may be added to the lyophilized formulation, pre-lyophilized formulation and/or the reconstituted formulation.
  • compositions may be included in the formulation (and/or the lyophilized formulation and/or the reconstituted formulation) provided that they do not adversely affect the desired characteristics of the formulation.
  • Acceptable carriers, excipients or stabilizers are nontoxic to recipients at the dosages and concentrations employed and include, but are not limited to, additional buffering agents; preservatives; co-solvents; antioxidants including ascorbic acid and methionine; chelating agents such as EDTA; metal complexes (e.g., Zn-protein complexes); biodegradable polymers such as polyesters; and/or salt-forming counterions such as sodium.
  • Formulations in either aqueous, pre-lyophilized, lyophilized or reconstituted form, in accordance with the present invention can be assessed based on product quality analysis, reconstitution time (if lyophilized), quality of reconstitution (if lyophilized), high molecular weight, moisture, and glass transition temperature.
  • protein quality and product analysis include product degradation rate analysis using methods including, but not limited to, size exclusion HPLC (SE-HPLC), cation exchange-HPLC (CEX-HPLC), X-ray diffraction (XRD), modulated differential scanning calorimetry (mDSC), reversed phase HPLC (RP-HPLC), multi-angle light scattering (MALS), fluorescence, ultraviolet absorption, nephelometry, capillary electrophoresis (CE), SDS-PAGE, and combinations thereof.
  • SE-HPLC size exclusion HPLC
  • CEX-HPLC cation exchange-HPLC
  • XRD X-ray diffraction
  • mDSC modulated differential scanning calorimetry
  • RP-HPLC reversed phase HPLC
  • MALS multi-angle light scattering
  • fluorescence ultraviolet absorption
  • nephelometry capillary electrophoresis
  • SDS-PAGE SDS-PAGE
  • evaluation of product in accordance with the present invention may include
  • formulations can be stored for extended periods of time at room temperature.
  • Storage temperature may typically range from 0° C. to 45° C. (e.g., 4° C., 20° C., 25° C., 45° C. etc.).
  • Formulations may be stored for a period of months to a period of years. Storage time generally will be 24 months, 12 months, 6 months, 4.5 months, 3 months, 2 months or 1 month. Formulations can be stored directly in the container used for administration, eliminating transfer steps.
  • Formulations can be stored directly in the lyophilization container (if lyophilized), which may also function as the reconstitution vessel, eliminating transfer steps. Alternatively, lyophilized product formulations may be measured into smaller increments for storage. Storage should generally avoid circumstances that lead to degradation of the proteins, including but not limited to exposure to sunlight, UV radiation, other forms of electromagnetic radiation, excessive heat or cold, rapid thermal shock, and mechanical shock.
  • Inventive methods in accordance with the present invention can be utilized to lyophilize any materials, in particular, therapeutic agents.
  • a pre-lyophilization formulation further contains an appropriate choice of excipients or other components such as stabilizers, buffering agents, bulking agents, and surfactants to prevent compound of interest from degradation (e.g., protein aggregation, deamidation, and/or oxidation) during freeze-drying and storage.
  • the formulation for lyophilization can include one or more additional ingredients including lyoprotectants or stabilizing agents, buffers, bulking agents, isotonicity agents and surfactants.
  • Lyophilization generally includes three main stages: freezing, primary drying and secondary drying. Freezing is necessary to convert water to ice or some amorphous formulation components to the crystalline form.
  • Primary drying is the process step when ice is removed from the frozen product by direct sublimation at low pressure and temperature.
  • Secondary drying is the process step when bounded water is removed from the product matrix utilizing the diffusion of residual water to the evaporation surface. Product temperature during secondary drying is normally higher than during primary drying. See, Tang X. et al. (2004) “Design of freeze-drying processes for pharmaceuticals: Practical advice,” Pharm. Res., 21:191-200; Nail S. L. et al.
  • an annealing step may be introduced during the initial freezing of the product.
  • the annealing step may reduce the overall cycle time. Without wishing to be bound by any theories, it is contemplated that the annealing step can help promote excipient crystallization and formation of larger ice crystals due to re-crystallization of small crystals formed during supercooling, which, in turn, improves reconstitution.
  • an annealing step includes an interval or oscillation in the temperature during freezing.
  • the freeze temperature may be ⁇ 40° C.
  • the annealing step will increase the temperature to, for example, ⁇ 10° C. and maintain this temperature for a set period of time.
  • the annealing step time may range from 0.5 hours to 8 hours (e.g., 0.5, 1.0 1.5, 2.0, 2.5, 3, 4, 6, and 8 hours).
  • the annealing temperature may be between the freezing temperature and 0° C.
  • Lyophilization may be performed in a container, such as a tube, a bag, a bottle, a tray, a vial (e.g., a glass vial), syringe or any other suitable containers.
  • the containers may be disposable. Lyophilization may also be performed in a large scale or small scale. In some instances, it may be desirable to lyophilize the protein formulation in the container in which reconstitution of the protein is to be carried out in order to avoid a transfer step.
  • the container in this instance may, for example, be a 3, 4, 5, 10, 20, 50 or 100 cc vial.
  • Freeze-drying is accomplished by freezing the formulation and subsequently subliming ice from the frozen content at a temperature suitable for primary drying. Initial freezing brings the formulation to a temperature below about ⁇ 20° C. (e.g., ⁇ 50° C., ⁇ 45° C., ⁇ 40° C., ⁇ 35° C., ⁇ 30° C., ⁇ 25° C., etc.) in typically not more than about 4 hours (e.g., not more than about 3 hours, not more than about 2.5 hours, not more than about 2 hours).
  • the product temperature is typically below the eutectic point or the collapse temperature of the formulation.
  • the shelf temperature for the primary drying will range from about ⁇ 30 to 25° C. (provided the product remains below the melting point during primary drying) at a suitable pressure, ranging typically from about 20 to 250 mTorr.
  • the formulation, size and type of the container holding the sample (e.g., glass vial) and the volume of liquid will mainly dictate the time required for drying, which can range from a few hours to several days.
  • a secondary drying stage is carried out at about 0-60° C., depending primarily on the type and size of container and the type of therapeutic protein employed. Again, volume of liquid will mainly dictate the time required for drying, which can range from a few hours to several days.
  • lyophilization will result in a lyophilized formulation in which the moisture content thereof is less than about 5%, less than about 4%, less than about 3%, less than about 2%, less than about 1%, and less than about 0.5%.
  • compositions of the present invention are generally in an aqueous form upon administration to a subject
  • the pharmaceutical compositions of the present invention are lyophilized.
  • Such compositions must be reconstituted by adding one or more diluents thereto prior to administration to a subject.
  • the lyophilized formulation may be reconstituted with a diluent such that the protein concentration in the reconstituted formulation is desirable.
  • a suitable diluent for reconstitution is water.
  • the water used as the diluent can be treated in a variety of ways including reverse osmosis, distillation, deionization, filtrations (e.g., activated carbon, microfiltration, nanofiltration) and combinations of these treatment methods.
  • the water should be suitable for injection including, but not limited to, sterile water or bacteriostatic water for injection.
  • Additional exemplary diluents include a pH buffered solution (e.g., phosphate-buffered saline), sterile saline solution, Elliot's solution, Ringer's solution or dextrose solution.
  • Suitable diluents may optionally contain a preservative.
  • Exemplary preservatives include aromatic alcohols such as benzyl or phenol alcohol. The amount of preservative employed is determined by assessing different preservative concentrations for compatibility with the protein and preservative efficacy testing. For example, if the preservative is an aromatic alcohol (such as benzyl alcohol), it can be present in an amount from about 0.1-2.0%, from about 0.5-1.5%, or about 1.0-1.2%.
  • Diluents suitable for the invention may include a variety of additives, including, but not limited to, pH buffering agents, (e.g. Tris, histidine) salts (e.g., sodium chloride) and other additives (e.g., sucrose) including those described above (e.g. stabilizing agents, isotonicity agents).
  • pH buffering agents e.g. Tris, histidine
  • salts e.g., sodium chloride
  • a lyophilized substance e.g., protein
  • a concentration of at least 25 mg/ml e.g., at least 50 mg/ml, at least 75 mg/ml, at least 100 mg
  • at least 25 mg/ml e.g., at least 50 mg/ml, at least 75 mg/ml, at least 100 mg
  • a lyophilized substance e.g., protein
  • a concentration ranging from about 1 mg/ml to 100 mg/ml (e.g., from about 1 mg/ml to 50 mg/ml, from 1 mg/ml to 100 mg/ml, from about 1 mg/ml to about 5 mg/ml, from about 1 mg/ml to about 10 mg/ml, from about 1 mg/ml to about 25 mg/ml, from about 1 mg/ml to about 75 mg/ml, from about 10 mg/ml to about 30 mg/ml, from about 10 mg/ml to about 50 mg/ml, from about 10 mg/ml to about 75 mg/ml, from about 10 mg/ml to about 100 mg/ml, from about 25 mg/ml to about 50 mg/ml, from about 25 mg/ml to about 75 mg/ml, from about 25 mg/ml to about 100 mg/ml, from about 50 mg/ml to about 75 mg/m/m
  • the concentration of protein in the reconstituted formulation may be higher than the concentration in the pre-lyophilization formulation.
  • High protein concentrations in the reconstituted formulation are considered to be particularly useful where subcutaneous or intramuscular delivery of the reconstituted formulation is intended.
  • the protein concentration in the reconstituted formulation may be about 2-50 times (e.g., about 2-20, about 2-10 times, or about 2-5 times) of the pre-lyophilized formulation.
  • the protein concentration in the reconstituted formulation may be at least about 2 times (e.g., at least about 3, 4, 5, 10, 20, 40 times) of the pre-lyophilized formulation.
  • Reconstitution according to the present invention may be performed in any container.
  • Exemplary containers suitable for the invention include, but are not limited to, such as tubes, vials, syringes (e.g., single-chamber or dual-chamber), bags, bottles, and trays.
  • Suitable containers may be made of any materials such as glass, plastics, metal.
  • the containers may be disposable or reusable. Reconstitution may also be performed in a large scale or small scale.
  • a suitable container for lyophilization and reconstitution is a dual chamber syringe (e.g., Lyo-Ject,® (Vetter) syringes).
  • a dual chamber syringe may contain both the lyophilized substance and the diluent, each in a separate chamber, separated by a stopper (see Example 5).
  • a plunger can be attached to the stopper at the diluent side and pressed to move diluent into the product chamber so that the diluent can contact the lyophilized substance and reconstitution may take place as described herein (see Example 5).
  • the pharmaceutical compositions, formulations and related methods of the invention are useful for delivering a variety of therapeutic agents to the CNS of a subject (e.g., intrathecally, intraventricularly or intracisternally) and for the treatment of the associated diseases.
  • the pharmaceutical compositions of the present invention are particularly useful for delivering proteins and enzymes (e.g., enzyme replacement therapy) to subjects suffering from lysosomal storage disorders.
  • the lysosomal storage diseases represent a group of relatively rare inherited metabolic disorders that result from defects in lysosomal function.
  • the lysosomal diseases are characterized by the accumulation of undigested macromolecules within the lysosomes, which results in an increase in the size and number of such lysosomes and ultimately in cellular dysfunction and clinical abnormalities.
  • Stable formulations described herein are generally suitable for CNS delivery of therapeutic agents.
  • Stable formulations according to the present invention can be used for CNS delivery via various techniques and routes including, but not limited to, intraparenchymal, intracerebral, intravetricular cerebral (ICV), intrathecal (e.g., IT-Lumbar, IT-cisterna magna) administrations and any other techniques and routes for injection directly or indirectly to the CNS and/or CSF.
  • ICV intravetricular cerebral
  • intrathecal e.g., IT-Lumbar, IT-cisterna magna
  • a replacement enzyme is delivered to the CNS in a formulation described herein.
  • a replacement enzyme is delivered to the CNS by administering into the cerebrospinal fluid (CSF) of a subject in need of treatment.
  • intrathecal administration is used to deliver a desired replacement enzyme (e.g., an ASA protein) into the CSF.
  • a desired replacement enzyme e.g., an ASA protein
  • intrathecal administration refers to an injection into the spinal canal (intrathecal space surrounding the spinal cord).
  • Various techniques may be used including, without limitation, lateral cerebroventricular injection through a burrhole or cistemal or lumbar puncture or the like. Exemplary methods are described in Lazorthes et al. Advances in Drug Delivery Systems and Applications in Neurosurgery, 143-192 and Omaya et al., Cancer Drug Delivery, 1: 169-179, the contents of which are incorporated herein by reference.
  • an enzyme may be injected at any region surrounding the spinal canal.
  • an enzyme is injected into the lumbar area or the cisterna magna or intraventricularly into a cerebral ventricle space.
  • the term “lumbar region” or “lumbar area” refers to the area between the third and fourth lumbar (lower back) vertebrae and, more inclusively, the L2-S1 region of the spine.
  • intrathecal injection via the lumbar region or lumber area is also referred to as “lumbar IT delivery” or “lumbar IT administration.”
  • cisterna magna refers to the space around and below the cerebellum via the opening between the skull and the top of the spine.
  • intrathecal injection via cisterna magna is also referred to as “cisterna magna delivery.”
  • Cerebral ventricle refers to the cavities in the brain that are continuous with the central canal of the spinal cord.
  • injections via the cerebral ventricle cavities are referred to as intravetricular Cerebral (ICV) delivery.
  • intrathecal administration or “intrathecal delivery” according to the present invention refers to lumbar IT administration or delivery, for example, delivered between the third and fourth lumbar (lower back) vertebrae and, more inclusively, the L2-S1 region of the spine. It is contemplated that lumbar IT administration or delivery distinguishes over cisterna magna delivery in that lumbar IT administration or delivery according to our invention provides better and more effective delivery to the distal spinal canal, while cisterna magna delivery, among other things, typically does not deliver well to the distal spinal canal.
  • a device for intrathecal administration contains a fluid access port (e.g., injectable port); a hollow body (e.g., catheter) having a first flow orifice in fluid communication with the fluid access port and a second flow orifice configured for insertion into spinal cord; and a securing mechanism for securing the insertion of the hollow body in the spinal cord.
  • a suitable securing mechanism contains one or more nobs mounted on the surface of the hollow body and a sutured ring adjustable over the one or more nobs to prevent the hollow body (e.g., catheter) from slipping out of the spinal cord.
  • the fluid access port comprises a reservoir.
  • the fluid access port comprises a mechanical pump (e.g., an infusion pump).
  • an implanted catheter is connected to either a reservoir (e.g., for bolus delivery), or an infusion pump.
  • the fluid access port may be implanted or external
  • intrathecal administration may be performed by either lumbar puncture (i.e., slow bolus) or via a port-catheter delivery system (i.e., infusion or bolus).
  • the catheter is inserted between the laminae of the lumbar vertebrae and the tip is threaded up the thecal space to the desired level (generally L3-L4) ( FIG. 56C ).
  • a single dose volume suitable for intrathecal administration is typically small.
  • intrathecal delivery according to the present invention maintains the balance of the composition of the CSF as well as the intracranial pressure of the subject.
  • intrathecal delivery is performed absent the corresponding removal of CSF from a subject.
  • a suitable single dose volume may be e.g., less than about 10 ml, 8 ml, 6 ml, 5 ml, 4 ml, 3 ml, 2 ml, 1.5 ml, 1 ml, or 0.5 ml.
  • a suitable single dose volume may be about 0.5-5 ml, 0.5-4 ml, 0.5-3 ml, 0.5-2 ml, 0.5-1 ml, 1-3 ml, 1-5 ml, 1.5-3 ml, 1-4 ml, or 0.5-1.5 ml.
  • intrathecal delivery according to the present invention involves a step of removing a desired amount of CSF first.
  • less than about 10 ml e.g., less than about 9 ml, 8 ml, 7 ml, 6 ml, 5 ml, 4 ml, 3 ml, 2 ml, 1 ml
  • a suitable single dose volume may be e.g., more than about 3 ml, 4 ml, 5 ml, 6 ml, 7 ml, 8 ml, 9 ml, 10 ml, 15 ml, or 20 ml.
  • a ventricular tube is inserted through a hole formed in the anterior horn and is connected to an Ommaya reservoir installed under the scalp, and the reservoir is subcutaneously punctured to intrathecally deliver the particular enzyme being replaced, which is injected into the reservoir.
  • the drug may be intrathecally given, for example, by a single injection, or continuous infusion. It should be understood that the dosage treatment may be in the form of a single dose administration or multiple doses.
  • formulations of the invention can be formulated in liquid solutions.
  • the enzyme may be formulated in solid form and re-dissolved or suspended immediately prior to use. Lyophilized forms are also included.
  • the injection can be, for example, in the form of a bolus injection or continuous infusion (e.g., using infusion pumps) of the enzyme.
  • the enzyme is administered by lateral cerebro ventricular injection into the brain of a subject.
  • the injection can be made, for example, through a burr hole made in the subject's skull.
  • the enzyme and/or other pharmaceutical formulation is administered through a surgically inserted shunt into the cerebral ventricle of a subject.
  • the injection can be made into the lateral ventricles, which are larger.
  • injection into the third and fourth smaller ventricles can also be made.
  • compositions used in the present invention are administered by injection into the cisterna magna, or lumbar area of a subject.
  • the pharmaceutically acceptable formulation provides sustained delivery, e.g., “slow release” of the enzyme or other pharmaceutical composition used in the present invention, to a subject for at least one, two, three, four weeks or longer periods of time after the pharmaceutically acceptable formulation is administered to the subject.
  • sustained delivery e.g., “slow release” of the enzyme or other pharmaceutical composition used in the present invention
  • sustained delivery refers to continual delivery of a pharmaceutical formulation of the invention in vivo over a period of time following administration, preferably at least several days, a week or several weeks.
  • Sustained delivery of the composition can be demonstrated by, for example, the continued therapeutic effect of the enzyme over time (e.g., sustained delivery of the enzyme can be demonstrated by continued reduced amount of storage granules in the subject).
  • sustained delivery of the enzyme may be demonstrated by detecting the presence of the enzyme in vivo over time.
  • therapeutic agents in particular, replacement enzymes administered using inventive methods and compositions of the present invention are able to effectively and extensively diffuse across the brain surface and penetrate various layers or regions of the brain, including deep brain regions.
  • inventive methods and compositions of the present invention effectively deliver therapeutic agents (e.g., an ASA enzyme) to various tissues, neurons or cells of spinal cord, including the lumbar region, which is hard to target by existing CNS delivery methods such as ICV injection.
  • inventive methods and compositions of the present invention deliver sufficient amount of therapeutic agents (e.g., an ASA enzyme) to blood stream and various peripheral organs and tissues.
  • a therapeutic protein e.g., an ASA enzyme
  • a therapeutic protein is delivered to the central nervous system of a subject.
  • a therapeutic protein e.g., an ASA enzyme
  • a therapeutic protein is delivered to one or more of target tissues of brain, spinal cord, and/or peripheral organs.
  • target tissues refers to any tissue that is affected by the lysosomal storage disease to be treated or any tissue in which the deficient lysosomal enzyme is normally expressed.
  • target tissues include those tissues in which there is a detectable or abnormally high amount of enzyme substrate, for example stored in the cellular lysosomes of the tissue, in patients suffering from or susceptible to the lysosomal storage disease.
  • target tissues include those tissues that display disease-associated pathology, symptom, or feature.
  • target tissues include those tissues in which the deficient lysosomal enzyme is normally expressed at an elevated level.
  • a target tissue may be a brain target tissue, a spinal cord target tissue and/or a peripheral target tissue. Exemplary target tissues are described in detail below.
  • meningeal tissue is a system of membranes which envelops the central nervous system, including the brain.
  • the meninges contain three layers, including dura matter, arachnoid matter, and pia matter.
  • the primary function of the meninges and of the cerebrospinal fluid is to protect the central nervous system.
  • a therapeutic protein in accordance with the present invention is delivered to one or more layers of the meninges.
  • the brain has three primary subdivisions, including the cerebrum, cerebellum, and brain stem.
  • the cerebral hemispheres which are situated above most other brain structures and are covered with a cortical layer. Underneath the cerebrum lies the brainstem, which resembles a stalk on which the cerebrum is attached.
  • the cerebellum At the rear of the brain, beneath the cerebrum and behind the brainstem, is the cerebellum.
  • the diencephalon which is located near the midline of the brain and above the mesencephalon, contains the thalamus, metathalamus, hypothalamus, epithalamus, prethalamus, and pretectum.
  • the mesencephalon also called the midbrain, contains the tectum, tegumentum, ventricular mesocoelia, and cerebral peduncels, the red nucleus, and the cranial nerve III nucleus.
  • the mesencephalon is associated with vision, hearing, motor control, sleep/wake, alertness, and temperature regulation.
  • Regions of tissues of the central nervous system, including the brain, can be characterized based on the depth of the tissues.
  • CNS e.g., brain
  • tissues can be characterized as surface or shallow tissues, mid-depth tissues, and/or deep tissues.
  • a therapeutic protein may be delivered to any appropriate brain target tissue(s) associated with a particular disease to be treated in a subject.
  • a therapeutic protein e.g., a replacement enzyme
  • a therapeutic protein in accordance with the present invention is delivered to surface or shallow brain target tissue.
  • a therapeutic protein in accordance with the present invention is delivered to mid-depth brain target tissue.
  • a therapeutic protein in accordance with the present invention is delivered to deep brain target tissue.
  • a therapeutic protein in accordance with the present invention is delivered to a combination of surface or shallow brain target tissue, mid-depth brain target tissue, and/or deep brain target tissue.
  • a therapeutic protein in accordance with the present invention is delivered to a deep brain tissue at least 4 mm, 5 mm, 6 mm, 7 mm, 8 mm, 9 mm, 10 mm or more below (or internal to) the external surface of the brain.
  • therapeutic agents are delivered to one or more surface or shallow tissues of cerebrum.
  • the targeted surface or shallow tissues of the cerebrum are located within 4 mm from the surface of the cerebrum.
  • the targeted surface or shallow tissues of the cerebrum are selected from pia mater tissues, cerebral cortical ribbon tissues, hippocampus, Virchow Robin space, blood vessels within the VR space, the hippocampus, portions of the hypothalamus on the inferior surface of the brain, the optic nerves and tracts, the olfactory bulb and projections, and combinations thereof.
  • therapeutic agents are delivered to one or more deep tissues of the cerebrum.
  • the targeted surface or shallow tissues of the cerebrum are located 4 mm (e.g., 5 mm, 6 mm, 7 mm, 8 mm, 9 mm, or 10 mm) below (or internal to) the surface of the cerebrum.
  • targeted deep tissues of the cerebrum include the cerebral cortical ribbon.
  • targeted deep tissues of the cerebrum include one or more of the diencephalon (e.g., the hypothalamus, thalamus, prethalamus, subthalamus, etc.), metencephalon, lentiform nuclei, the basal ganglia, caudate, putamen, amygdala, globus pallidus, and combinations thereof.
  • diencephalon e.g., the hypothalamus, thalamus, prethalamus, subthalamus, etc.
  • metencephalon e.g., the hypothalamus, thalamus, prethalamus, subthalamus, etc.
  • metencephalon e.g., metencephalon
  • lentiform nuclei e.g., the basal ganglia, caudate, putamen, amygdala, globus pallidus, and combinations thereof.
  • therapeutic agents are delivered to one or more tissues of the cerebellum.
  • the targeted one or more tissues of the cerebellum are selected from the group consisting of tissues of the molecular layer, tissues of the Purkinje cell layer, tissues of the Granular cell layer, cerebellar peduncles, and combination thereof.
  • therapeutic agents e.g., enzymes
  • therapeutic agents are delivered to one or more tissues of the brainstem.
  • the targeted one or more tissues of the brainstem include brain stem white matter tissue and/or brain stem nuclei tissue.
  • therapeutic agents are delivered to various brain tissues including, but not limited to, gray matter, white matter, periventricular areas, pia-arachnoid, meninges, neocortex, cerebellum, deep tissues in cerebral cortex, molecular layer, caudate/putamen region, midbrain, deep regions of the pons or medulla, and combinations thereof.
  • therapeutic agents are delivered to various cells in the brain including, but not limited to, neurons, glial cells, perivascular cells and/or meningeal cells.
  • a therapeutic protein is delivered to oligodendrocytes of deep white matter.
  • regions or tissues of the spinal cord can be characterized based on the depth of the tissues.
  • spinal cord tissues can be characterized as surface or shallow tissues, mid-depth tissues, and/or deep tissues.
  • therapeutic agents are delivered to one or more surface or shallow tissues of the spinal cord.
  • a targeted surface or shallow tissue of the spinal cord is located within 4 mm from the surface of the spinal cord.
  • a targeted surface or shallow tissue of the spinal cord contains pia matter and/or the tracts of white matter.
  • therapeutic agents are delivered to one or more deep tissues of the spinal cord.
  • a targeted deep tissue of the spinal cord is located internal to 4 mm from the surface of the spinal cord.
  • a targeted deep tissue of the spinal cord contains spinal cord grey matter and/or ependymal cells.
  • therapeutic agents e.g., enzymes
  • neurons of the spinal cord are delivered to therapeutic agents.
  • peripheral organs or tissues refer to any organs or tissues that are not part of the central nervous system (CNS).
  • Peripheral target tissues may include, but are not limited to, blood system, liver, kidney, heart, endothelium, bone marrow and bone marrow derived cells, spleen, lung, lymph node, bone, cartilage, ovary and testis.
  • a therapeutic protein e.g., a replacement enzyme in accordance with the present invention is delivered to one or more of the peripheral target tissues.
  • a therapeutic agent e.g., an ASA enzyme
  • a therapeutic agent e.g., enzyme
  • a target cell e.g., neurons such as Purkinje cells
  • intrathecally-administered enzymes demonstrate translocation dynamics such that the enzyme moves within the perivascular space (e.g., by pulsation-assisted convective mechanisms).
  • active axonal transport mechanisms relating to the association of the administered protein or enzyme with neurofilaments may also contribute to or otherwise facilitate the distribution of intrathecally-administered proteins or enzymes into the deeper tissues of the central nervous system.
  • a therapeutic agent e.g., an ASA enzyme delivered according to the present invention may achieve therapeutically or clinically effective levels or activities in various targets tissues described herein.
  • a therapeutically or clinically effective level or activity is a level or activity sufficient to confer a therapeutic effect in a target tissue.
  • the therapeutic effect may be objective (i.e., measurable by some test or marker) or subjective (i.e., subject gives an indication of or feels an effect).
  • a therapeutically or clinically effective level or activity may be an enzymatic level or activity that is sufficient to ameliorate symptoms associated with the disease in the target tissue (e.g., GAG storage).
  • a therapeutic agent (e.g., a replacement enzyme) delivered according to the present invention may achieve an enzymatic level or activity that is at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% of the normal level or activity of the corresponding lysosomal enzyme in the target tissue.
  • a therapeutic agent (e.g., a replacement enzyme) delivered according to the present invention may achieve an enzymatic level or activity that is increased by at least 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold or 10-fold as compared to a control (e.g., endogenous levels or activities without the treatment).
  • a therapeutic agent e.g., a replacement enzyme delivered according to the present invention may achieve an increased enzymatic level or activity at least approximately 10 nmol/hr/mg, 20 nmol/hr/mg, 40 nmol/hr/mg, 50 nmol/hr/mg, 60 nmol/hr/mg, 70 nmol/hr/mg, 80 nmol/hr/mg, 90 nmol/hr/mg, 100 nmol/hr/mg, 150 nmol/hr/mg, 200 nmol/hr/mg, 250 nmol/hr/mg, 300 nmol/hr/mg, 350 nmol/hr/mg, 400 nmol/hr/mg, 450 nmol/hr/mg, 500 nmol/hr/mg, 550 nmol/hr/mg or 600 nmol/hr/mg in a target tissue
  • inventive methods according to the present invention are particularly useful for targeting the lumbar region.
  • a therapeutic agent e.g., a replacement enzyme
  • a therapeutic agent delivered according to the present invention may achieve an increased enzymatic level or activity in the lumbar region of at least approximately 500 nmol/hr/mg, 600 nmol/hr/mg, 700 nmol/hr/mg, 800 nmol/hr/mg, 900 nmol/hr/mg, 1000 nmol/hr/mg, 1500 nmol/hr/mg, 2000 nmol/hr/mg, 3000 nmol/hr/mg, 4000 nmol/hr/mg, 5000 nmol/hr/mg, 6000 nmol/hr/mg, 7000 nmol/hr/mg, 8000 nmol/hr/mg, 9000 nmol/hr/mg, or 10,000 nmol/hr/mg.
  • therapeutic agents e.g., replacement enzymes
  • a therapeutic agent e.g., a replacement enzyme
  • a therapeutic agent delivered according to the present invention may have a half-life of at least approximately 30 minutes, 45 minutes, 60 minutes, 90 minutes, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 12 hours, 16 hours, 18 hours, 20 hours, 25 hours, 30 hours, 35 hours, 40 hours, up to 3 days, up to 7 days, up to 14 days, up to 21 days or up to a month.
  • a therapeutic agent e.g., a replacement enzyme delivered according to the present invention may retain detectable level or activity in CSF or bloodstream after 12 hours, 24 hours, 30 hours, 36 hours, 42 hours, 48 hours, 54 hours, 60 hours, 66 hours, 72 hours, 78 hours, 84 hours, 90 hours, 96 hours, 102 hours, or a week following administration. Detectable level or activity may be determined using various methods known in the art.
  • a therapeutic agent e.g., a replacement enzyme delivered according to the present invention achieves a concentration of at least 30 ⁇ g/ml in the CNS tissues and cells of the subject following administration (e.g., one week, 3 days, 48 hours, 36 hours, 24 hours, 18 hours, 12 hours, 8 hours, 6 hours, 4 hours, 3 hours, 2 hours, 1 hour, 30 minutes, or less, following intrathecal administration of the pharmaceutical composition to the subject).
  • a therapeutic agent e.g., a replacement enzyme delivered according to the present invention achieves a concentration of at least 20 ⁇ g/ml, at least 15 ⁇ g/ml, at least 10 ⁇ g/ml, at least 7.5 ⁇ g/ml, at least 5 ⁇ g/ml, at least 2.5 ⁇ g/ml, at least 1.0 ⁇ g/ml or at least 0.5 ⁇ g/ml in the targeted tissues or cells of the subject (e.g., brain tissues or neurons) following administration to such subject (e.g., one week, 3 days, 48 hours, 36 hours, 24 hours, 18 hours, 12 hours, 8 hours, 6 hours, 4 hours, 3 hours, 2 hours, 1 hour, 30 minutes, or less following intrathecal administration of such pharmaceutical compositions to the subject).
  • a therapeutic agent e.g., a replacement enzyme
  • MLD Metachromatic Leukodystrophy Disease
  • Metachromatic Leukodystrophy Disease is an autosomal recessive disorder resulting from a deficiency of the enzyme Arylsulfatease A (ASA).
  • ASA which is encoded by the ARSA gene in humans, is an enzyme that breaks down cerebroside 3-sulfate or sphingolipid 3-O-sulfogalactosylceramide (sulfatide) into cerebroside and sulfate.
  • sulfatides accumulate in the nervous system (e.g., myelin sheaths, neurons and glial cells) and to a lesser extent in visceral organs. The consequence of these molecular and cellular events is progressive demyelination and axonal loss within the CNS and PNS, which is accompanied clinically by severe motor and cognitive dysfunction.
  • a defining clinical feature of this disorder is central nervous system (CNS) degeneration, which results in cognitive impairment (e.g., mental retardation, nervous disorders, and blindness, among others).
  • CNS central nervous system
  • MLD can manifest itself in young children (Late-infantile form), where affected children typically begin showing symptoms just after the first year of life (e.g., at about 15-24 months), and generally do not survive past the age of 5 years. MLD can manifest itself in children (Juvenile form), where affected children typically show cognitive impairment by about the age of 3-10 years, and life-span can vary (e.g., in the range of 10-15 years after onset of symptoms). MLD can manifest itself in adults (Adult-onset form) and can appear in individuals of any age (e.g., typically at age 16 and later) and the progression of the disease can vary greatly.
  • compositions and methods of the present invention may be used to effectively treat individuals suffering from or susceptible to MLD.
  • treat or “treatment,” as used herein, refers to amelioration of one or more symptoms associated with the disease, prevention or delay of the onset of one or more symptoms of the disease, and/or lessening of the severity or frequency of one or more symptoms of the disease.
  • Exemplary symptoms include, but are not limited to, intracranial pressure, hydrocephalus ex vacuo, accumulated sulfated glycolipids in the myelin sheaths in the central and peripheral nervous system and in visceral organs, progressive demyelination and axonal loss within the CNS and PNS, and/or motor and cognitive dysfunction.
  • treatment refers to partially or complete alleviation, amelioration, relief, inhibition, delaying onset, reducing severity and/or incidence of neurological impairment in an MLD patient.
  • neurological impairment includes various symptoms associated with impairment of the central nervous system (e.g., the brain and spinal cord).
  • various symptoms of MLD are associated with impairment of the peripheral nervous system (PNS).
  • PNS peripheral nervous system
  • neurological impairment in an MLD patient is characterized by decline in gross motor function. It will be appreciated that gross motor function may be assessed by any appropriate method. For example, in some embodiments, gross motor function is measured as the change from a baseline in motor function using the Gross Motor Function Measure-88 (GMFM-88) total raw score.
  • GMFM-88 Gross Motor Function Measure-88
  • treatment refers to decreased sulfatide accumulation in various tissues. In some embodiments, treatment refers to decreased sulfatide accumulation in brain target tissues, spinal cord neurons, and/or peripheral target tissues. In certain embodiments, sulfatide accumulation is decreased by about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100% or more as compared to a control. In some embodiments, sulfatide accumulation is decreased by at least 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold or 10-fold as compared to a control.
  • sulfatide storage may be assessed by any appropriate method.
  • sulfatide storage is measured by alcian blue staining.
  • sulfatide storage is measured by LAMP-1 staining.
  • treatment refers to reduced vacuolization in neurons (e.g., neurons containing Purkinje cells).
  • vacuolization in neurons is decreased by about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100% or more as compared to a control.
  • vacuolization is decreased by at least 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold or 10-fold as compared to a control.
  • treatment refers to increased ASA enzyme activity in various tissues.
  • treatment refers to increased ASA enzyme activity in brain target tissues, spinal cord neurons and/or peripheral target tissues.
  • ASA enzyme activity is increased by about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900% 1000% or more as compared to a control.
  • ASA enzyme activity is increased by at least 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold or 10-fold as compared to a control.
  • increased ASA enzymatic activity is at least approximately 10 nmol/hr/mg, 20 nmol/hr/mg, 40 nmol/hr/mg, 50 nmol/hr/mg, 60 nmol/hr/mg, 70 nmol/hr/mg, 80 nmol/hr/mg, 90 nmol/hr/mg, 100 nmol/hr/mg, 150 nmol/hr/mg, 200 nmol/hr/mg, 250 nmol/hr/mg, 300 nmol/hr/mg, 350 nmol/hr/mg, 400 nmol/hr/mg, 450 nmol/hr/mg, 500 nmol/hr
  • ASA enzymatic activity is increased in the lumbar region.
  • increased ASA enzymatic activity in the lumbar region is at least approximately 2000 nmol/hr/mg, 3000 nmol/hr/mg, 4000 nmol/hr/mg, 5000 nmol/hr/mg, 6000 nmol/hr/mg, 7000 nmol/hr/mg, 8000 nmol/hr/mg, 9000 nmol/hr/mg, 10,000 nmol/hr/mg, or more.
  • treatment refers to decreased progression of loss of cognitive ability. In certain embodiments, progression of loss of cognitive ability is decreased by about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%. 85%, 90%, 95%, 100% or more as compared to a control. In some embodiments, treatment refers to decreased developmental delay. In certain embodiments, developmental delay is decreased by about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 100% or more as compared to a control.
  • treatment refers to increased survival (e.g. survival time).
  • treatment can result in an increased life expectancy of a patient.
  • treatment according to the present invention results in an increased life expectancy of a patient by more than about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 105%, about 110%, about 115%, about 120%, about 125%, about 130%, about 135%, about 140%, about 145%, about 150%, about 155%, about 160%, about 165%, about 170%, about 175%, about 180%, about 185%, about 190%, about 195%, about 200% or more, as compared to the average life expectancy of one or more control individuals with similar disease without treatment.
  • treatment according to the present invention results in an increased life expectancy of a patient by more than about 6 month, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 12 months, about 2 years, about 3 years, about 4 years, about 5 years, about 6 years, about 7 years, about 8 years, about 9 years, about 10 years or more, as compared to the average life expectancy of one or more control individuals with similar disease without treatment.
  • treatment according to the present invention results in long term survival of a patient.
  • the term “long term survival” refers to a survival time or life expectancy longer than about 40 years, 45 years, 50 years, 55 years, 60 years, or longer.
  • a suitable control is a baseline measurement, such as a measurement in the same individual prior to initiation of the treatment described herein, or a measurement in a control individual (or multiple control individuals) in the absence of the treatment described herein.
  • a “control individual” is an individual afflicted with the same form MLD (e.g., late-infantile, juvenile, or adult-onset form), who is about the same age and/or gender as the individual being treated (to ensure that the stages of the disease in the treated individual and the control individual(s) are comparable).
  • the individual (also referred to as “patient” or “subject”) being treated is an individual (fetus, infant, child, adolescent, or adult human) having MLD or having the potential to develop MLD.
  • the individual can have residual endogenous ASA expression and/or activity, or no measurable activity.
  • the individual having MLD may have ASA expression levels that are less than about 30-50%, less than about 25-30%, less than about 20-25%, less than about 15-20%, less than about 10-15%, less than about 5-10%, less than about 0.1-5% of normal ASA expression levels.
  • the individual is an individual who has been recently diagnosed with the disease.
  • early treatment treatment commencing as soon as possible after diagnosis
  • intrathecal administration of a therapeutic agent does not result in severe adverse effects in the subject.
  • severe adverse effects induce, but are not limited to, substantial immune response, toxicity, or death.
  • substantial immune response refers to severe or serious immune responses, such as adaptive T-cell immune responses.
  • inventive methods according to the present invention do not involve concurrent immunosuppressant therapy (i.e., any immunosuppressant therapy used as pre-treatment/pre-conditioning or in parallel to the method).
  • inventive methods according to the present invention do not involve an immune tolerance induction in the subject being treated.
  • inventive methods according to the present invention do not involve a pre-treatment or preconditioning of the subject using T-cell immunosuppressive agent.
  • intrathecal administration of therapeutic agents can mount an immune response against these agents.
  • Immune tolerance may be induced using various methods known in the art. For example, an initial 30-60 day regimen of a T-cell immunosuppressive agent such as cyclosporin A (CsA) and an antiproliferative agent, such as, azathioprine (Aza), combined with weekly intrathecal infusions of low doses of a desired replacement enzyme may be used.
  • CsA cyclosporin A
  • an antiproliferative agent such as, azathioprine (Aza)
  • immunosuppressant agent any immunosuppressant agent known to the skilled artisan may be employed together with a combination therapy of the invention.
  • immunosuppressant agents include but are not limited to cyclosporine, FK506, rapamycin, CTLA4-Ig, and anti-TNF agents such as etanercept (see e.g. Moder, 2000, Ann. Allergy Asthma Immunol. 84, 280-284; Nevins, 2000, Curr. Opin. Pediatr. 12, 146-150; Kurlberg et al., 2000, Scand. J. Immunol. 51, 224-230; Ideguchi et al., 2000, Neuroscience 95, 217-226; Potter et al., 1999, Ann.
  • the anti-IL2 receptor (.alpha.-subunit) antibody daclizumab (e.g. ZenapaxTM), which has been demonstrated effective in transplant patients, can also be used as an immunosuppressant agent (see e.g.
  • Additional immunosuppressant agents include but are not limited to anti-CD2 (Branco et al., 1999, Transplantation 68, 1588-1596; Przepiorka et al., 1998, Blood 92, 4066-4071), anti-CD4 (Marinova-Mutafchieva et al., 2000, Arthritis Rheum. 43, 638-644; Fishwild et al., 1999, Clin. Immunol. 92, 138-152), and anti-CD40 ligand (Hong et al., 2000, Semin. Nephrol. 20, 108-125; Chirmule et al., 2000, J. Virol. 74, 3345-3352; Ito et al., 2000, J. Immunol. 164, 1230-1235).
  • Inventive methods of the present invention contemplate single as well as multiple administrations of a therapeutically effective amount of the therapeutic agents (e.g., replacement enzymes) described herein.
  • Therapeutic agents e.g., replacement enzymes
  • a therapeutically effective amount of the therapeutic agents (e.g., replacement enzymes) of the present invention may be administered intrathecally periodically at regular intervals (e.g., once every year, once every six months, once every five months, once every three months, bimonthly (once every two months), monthly (once every month), biweekly (once every two weeks), weekly).
  • intrathecal administration may be used in conjunction with other routes of administration (e.g., intravenous, subcutaneously, intramuscularly, parenterally, transdermally, or transmucosally (e.g., orally or nasally)).
  • those other routes of administration e.g., intravenous administration
  • a therapeutically effective amount is largely determined based on the total amount of the therapeutic agent contained in the pharmaceutical compositions of the present invention. Generally, a therapeutically effective amount is sufficient to achieve a meaningful benefit to the subject (e.g., treating, modulating, curing, preventing and/or ameliorating the underlying disease or condition).
  • a therapeutically effective amount may be an amount sufficient to achieve a desired therapeutic and/or prophylactic effect, such as an amount sufficient to modulate lysosomal enzyme receptors or their activity to thereby treat such lysosomal storage disease or the symptoms thereof (e.g., a reduction in or elimination of the presence or incidence of “zebra bodies” or cellular vacuolization following the administration of the compositions of the present invention to a subject).
  • a therapeutic agent e.g., a recombinant lysosomal enzyme
  • administered to a subject in need thereof will depend upon the characteristics of the subject. Such characteristics include the condition, disease severity, general health, age, sex and body weight of the subject.
  • characteristics include the condition, disease severity, general health, age, sex and body weight of the subject.
  • objective and subjective assays may optionally be employed to identify optimal dosage ranges.
  • a therapeutically effective amount is commonly administered in a dosing regimen that may comprise multiple unit doses.
  • a therapeutically effective amount (and/or an appropriate unit dose within an effective dosing regimen) may vary, for example, depending on route of administration, on combination with other pharmaceutical agents.
  • the specific therapeutically effective amount (and/or unit dose) for any particular patient may depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific pharmaceutical agent employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and/or rate of excretion or metabolism of the specific fusion protein employed; the duration of the treatment; and like factors as is well known in the medical arts.
  • the therapeutically effective dose ranges from about 0.005 mg/kg brain weight to 500 mg/kg brain weight, e.g., from about 0.005 mg/kg brain weight to 400 mg/kg brain weight, from about 0.005 mg/kg brain weight to 300 mg/kg brain weight, from about 0.005 mg/kg brain weight to 200 mg/kg brain weight, from about 0.005 mg/kg brain weight to 100 mg/kg brain weight, from about 0.005 mg/kg brain weight to 90 mg/kg brain weight, from about 0.005 mg/kg brain weight to 80 mg/kg brain weight, from about 0.005 mg/kg brain weight to 70 mg/kg brain weight, from about 0.005 mg/kg brain weight to 60 mg/kg brain weight, from about 0.005 mg/kg brain weight to 50 mg/kg brain weight, from about 0.005 mg/kg brain weight to 40 mg/kg brain weight, from about 0.005 mg/kg brain weight to 30 mg/kg brain weight, from about 0.005 mg/kg brain weight to 25 mg/kg brain weight, from about 0.005 mg/
  • the therapeutically effective dose is greater than about 0.1 mg/kg brain weight, greater than about 0.5 mg/kg brain weight, greater than about 1.0 mg/kg brain weight, greater than about 3 mg/kg brain weight, greater than about 5 mg/kg brain weight, greater than about 10 mg/kg brain weight, greater than about 15 mg/kg brain weight, greater than about 20 mg/kg brain weight, greater than about 30 mg/kg brain weight, greater than about 40 mg/kg brain weight, greater than about 50 mg/kg brain weight, greater than about 60 mg/kg brain weight, greater than about 70 mg/kg brain weight, greater than about 80 mg/kg brain weight, greater than about 90 mg/kg brain weight, greater than about 100 mg/kg brain weight, greater than about 150 mg/kg brain weight, greater than about 200 mg/kg brain weight, greater than about 250 mg/kg brain weight, greater than about 300 mg/kg brain weight, greater than about 350 mg/kg brain weight, greater than about 400 mg/kg brain weight, greater than about 450 mg/kg brain weight, greater than about 500 mg/kg brain weight.
  • the therapeutically effective dose may also be defined by mg/kg body weight.
  • the brain weights and body weights can be correlated. Dekaban A S. “Changes in brain weights during the span of human life: relation of brain weights to body heights and body weights,” Ann Neurol 1978; 4:345-56.
  • the dosages can be converted as shown in Table 5.
  • the therapeutically effective dose may also be defined by mg/15 cc of CSF.
  • therapeutically effective doses based on brain weights and body weights can be converted to mg/15 cc of CSF.
  • the volume of CSF in adult humans is approximately 150 mL (Johanson C E, et al. “Multiplicity of cerebrospinal fluid functions: New challenges in health and disease,” Cerebrospinal Fluid Res. 2008 May 14; 5:10). Therefore, single dose injections of 0.1 mg to 50 mg protein to adults would be approximately 0.01 mg/15 cc of CSF (0.1 mg) to 5.0 mg/15 cc of CSF (50 mg) doses in adults.
  • kits or other articles of manufacture which contains the formulation of the present invention and provides instructions for its reconstitution (if lyophilized) and/or use.
  • Kits or other articles of manufacture may include a container, an IDDD, a catheter and any other articles, devices or equipment useful in interthecal administration and associated surgery.
  • Suitable containers include, for example, bottles, vials, syringes (e.g., pre-filled syringes), ampules, cartridges, reservoirs, or lyo-jects.
  • the container may be formed from a variety of materials such as glass or plastic.
  • a container is a pre-filled syringe.
  • Suitable pre-filled syringes include, but are not limited to, borosilicate glass syringes with baked silicone coating, borosilicate glass syringes with sprayed silicone, or plastic resin syringes without silicone.
  • the container may holds formulations and a label on, or associated with, the container that may indicate directions for reconstitution and/or use.
  • the label may indicate that the formulation is reconstituted to protein concentrations as described above.
  • the label may further indicate that the formulation is useful or intended for, for example, IT administration.
  • a container may contain a single dose of a stable formulation containing a therapeutic agent (e.g., a replacement enzyme).
  • a single dose of the stable formulation is present in a volume of less than about 15 ml, 10 ml, 5.0 ml, 4.0 ml, 3.5 ml, 3.0 ml, 2.5 ml, 2.0 ml, 1.5 ml, 1.0 ml, or 0.5 ml.
  • a container holding the formulation may be a multi-use vial, which allows for repeat administrations (e.g., from 2-6 administrations) of the formulation.
  • Kits or other articles of manufacture may further include a second container comprising a suitable diluent (e.g., BWFI, saline, buffered saline).
  • the final protein concentration in the reconstituted formulation will generally be at least 1 mg/ml (e.g., at least 5 mg/ml, at least 10 mg/ml, at least 25 mg/ml, at least 50 mg/ml, at least 75 mg/ml, at least 100 mg/ml).
  • Kits or other articles of manufacture may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, IDDDs, catheters, syringes, and package inserts with instructions for use.
  • rhASA human arylsulfatase A
  • the concentrations of rhASA detected in the CNS tissues of the cynomolgus monkeys were analyzed by ELISA and compared to a therapeutic target of 10% of normal human rhASA concentrations, corresponding to approximately 2.5 ng/mg of tissue.
  • Tissue samples or punches were extracted from different areas of the brains of the cynomolgus monkeys and further analyzed for the presence of rhASA.
  • FIG. 24 illustrates the tissues from which the punches were extracted.
  • the punched tissue samples reflected an increase in the concentrations of rhASA, as reflected in FIGS. 25A-G , with a deposition gradient from the cerebral cortex to the deep white matter and deep gray matter.
  • Concentrations of rhASA detected using the same punch from both the IT and ICV routes of administration for six monkeys administered the 18.6 mg dose of rhASA are illustrated in FIGS. 26A-B .
  • the concentrations of rhASA detected in the deep white matter ( FIG. 25A ) and in the deep grey matter ( FIG. 26B ) brain tissues of adult and juvenile cynomolgus monkeys intrathecally-(IT) or intracerebroventricularly-(ICV) administered rhASA were comparable.
  • the punched tissue samples extracted from the brains of adult and juvenile cynomolgus monkeys were then analyzed to determine the concentrations of rhASA deposited in the extracted tissue sample, and to compare such concentrations to the therapeutic target concentration of 2.5 ng rhASA per mg protein (corresponding to 10% of the normal concentration of rhASA in a healthy subject).
  • the concentrations of rhASA deposited in the extracted tissue sample were then analyzed to determine the concentrations of rhASA deposited in the extracted tissue sample, and to compare such concentrations to the therapeutic target concentration of 2.5 ng rhASA per mg protein (corresponding to 10% of the normal concentration of rhASA in a healthy subject).
  • the 18.6 mg dose of IT-administered rhASA resulted in an rhASA concentration which exceeded the target therapeutic concentration of 2.5 ng/mg of protein.
  • each tissue sample punch analyzed demonstrated a concentration of rhASA either within or exceeding the therapeutic concentration of 2.5 ng/mg of protein and the median rhASA concentrations were above the therapeutic target for all tissue punches tested ( FIG. 27B ).
  • FIG. 28A Immunostaining of the deep white matter tissue revealed distribution of rhASA in the cynomolgus monkey in oligodendrocyte cells, as illustrated by FIG. 28B .
  • FIG. 28C illustrates that the IT-administered rhrASA demonstrated co-localization in the deep white matter tissues of the cynomolgus monkey. In particular, under staining co-localization in target organelles, such as the lysosome, is evident ( FIG.
  • rhASA labeled with the positron emitter 124 I was prepared and formulated in a vehicle of 154 mM NaCl, 0.005% polysorbate 20 at a pH of 6.0.
  • a volume of the formulation equivalent to 3 mg of rhASA (corresponding to approximately 38 mg/kg of brain) was administered to adult cynomolgus monkeys via intracerebroventricular (ICV) and intrathecal (IT) routes of administration.
  • ICV intracerebroventricular
  • IT intrathecal
  • the cynomolgus monkeys were subject to high-resolution PET scan imaging studies (microPET P4) to determine distribution of the administered 124 I-labeled rhASA.
  • PET imaging data ( FIG. 29 ) illustrates that both the ICV- and IT-administered 124 I-labeled rhASA effectively distributed to the tissues of the CNS, and in particular the 124 I-labeled rhASA administered through the IT-lumbar catheter immediately and uniformly spread in the cerebrospinal fluid (CSF) over the length of the spine.
  • CSF cerebrospinal fluid
  • therapeutic concentrations of 124 I-labeled rhASA were detected in the CNS tissues of the subject cynomolgus monkey, including the brain, spinal cord and CSF.
  • the concentrations of rhASA detected in such CNS tissues, and in particular in the tissues of the brain exceeded the therapeutic target concentration of 2.5 ng/mg of protein.
  • both the ICV- and IT-administered 124 I-labeled ASA effectively distributed to the tissues of the CNS.
  • twenty four hours following IT-administration 12.4% of the administered dose was in the cranial region, compared to 16.7% of the ICV-administered dose.
  • concentrations of rhASA detected in such CNS tissues, and in particular in the tissues of the brain, when rhASA was administered IT approached those concentrations detected following ICV-administration of the same dose.
  • ICV injection of the 124 I-labeled rhASA results ICV injection results in the immediate transfer of the injected volume to the cisterna magna, cisterna pontis, cisterna interpeduncularis and proximal spine, as illustrated in FIG. 30 .
  • within 2-5 hr IT administration delivered the 124 I-labeled rhASA to the same initial compartments (cisternae and proximal spine) as shown for the ICV administration. Twenty four hours following both ICV- and IT-administration distribution of the 124 I-labeled rhASA was comparable, as illustrated in FIG. 31 . Accordingly, unlike small molecules drugs, the foregoing results suggest that ICV-administration offers minimal advantages over IT-administration of rhASA.
  • the lysosomal storage diseases represent a family of genetic disorders caused by missing or defective enzymes which result in abnormal substrate accumulation. While the peripheral symptoms associated with several of these diseases can be effectively mitigated by intravenous administration of recombinant enzymes, intravenous administration of such recombinant enzymes are not expected to significantly impact the CNS manifestations associated with a majority of the lysosomal storage disease.
  • recombinant human iduronate-2-sulfatase (Idursulfase, Elaprase®; Shire Human Genetic Therapies, Inc. Lexington, Mass.) is approved for treatment of the somatic symptoms of Hunter syndrome but there is no pharmacologic therapy for the treatment of the neurologic manifestations which can include delayed development and progressive mental impairment. This is in part due to the nature of I2S, which is a large, highly-glycosylated enzyme with a molecular weight of approximately 76 kD and that does not traverse the blood brain barrier following intravenous administration.
  • the present inventors have therefore undertaken a program to investigate the intrathecal (IT) delivery of intrathecal formulations of recombinant human enzymes, such as, for example, iduronate-2-sulfatase (I2S), arylsulfatase A (rhASA) and alpha-N-acetylglucosaminidase (Naglu).
  • I2S iduronate-2-sulfatase
  • rhASA arylsulfatase A
  • Naglu alpha-N-acetylglucosaminidase
  • Immunohistochemical analyses of the CNS tissues demonstrated that the protein is targeted to the lysosome, the site of pathologic glycosaminoglycan accumulation in the lysosomal storage disorders. Furthermore, the morphologic improvements demonstrated in the IKO mouse model of Hunter syndrome, the Naglu-deficient mouse model of Sanfilippo syndrome type B, and the ASA knockout mouse model of metachromatic leukodystrohpy (MLD) reinforces the observation that IT-administered enzyme is distributed to the appropriate tissues and transported to the appropriate cellular compartments and organelles.
  • MLD metachromatic leukodystrohpy
  • Example 3 Formulations of Arylsulfatase A for it Administration
  • the stability data demonstrate that the saline formulation of drug substance and drug product (without PBS 20) is stable after 18 months at ⁇ 65 degrees C. and 18 months at 2-8 degrees C.
  • the solubility and stability of rhASA was investigated under limited buffer and excipient conditions due to its intended delivery to the CNS.
  • formulation development studies had been conducted to develop an intravenous (IV) formulation. Based on the results of these experiments, a formulation containing 30 mg/ml of rhASA in 10 mM citrate-phosphate buffer, pH 5.5 with 137 mM NaCl and 0.15% poloxomer 188 was selected as the lead IV formulation.
  • rhASA was also formulated for IT delivery in three formulations and stability data for this protein was investigated under these conditions. rhASA lots derived from upstream material product at one site were utilized. The results demonstrated that rhASA was stable in 154 mM sodium chloride solution with 0.005% polysorbate 20 (P20), pH 6.0 for at least 18 months at 2-8 degrees C. In addition, studies have been performed to demonstrate stability toward freeze-thaw and agitation-induced degradation.
  • rhASA formulated at 40 mg/mL rhASA in 10 mM citrate sodium phosphate with 137 mM NaCl, at pH 5.6 was dialyzed into five formulations which were utilized for IT preformulation studies (Table 8).
  • Tm melting temperature
  • DSC Differential Scanning calorimetry
  • a capillary DSC microcalorimeter (MicroCal) was employed at a scan rate of 60° C./hr and a temperature range of 10-110° C. Buffer baselines were subtracted from the protein scans. The scans were normalized for the protein concentration of each sample (measured by ultraviolet absorbance at 280 nm and using an extinction coefficient of 0.69 (mg/mL)-1.cm-1).
  • rhASA drug substance was subjected to either two weeks at 40° C. or one month at 40° C. Additional samples were placed on short term stability at 2-8° C. for 3 months. Samples were filtered (Millipore, P/N SLGV033RS) and aliquots of 0.5 mL were dispensed into 2 mL with 13 mm Flurotec stoppers.
  • Tm temperature midpoint of the thermally induces denaturation
  • Table 8 The effect of formulation composition (Table 8) on the Tm (temperature midpoint of the thermally induces denaturation) was investigated using DSC.
  • the Tm values for different formulation compositions are shown in FIG. 5 .
  • the Tm values exhibited similar unfolding temperatures for most of the formulations, except low Tm values were observed for rhASA formulated in either 5 mM sodium phosphate with 154 mM NaCl at pH 7.0 or 1 mM sodium phosphate with 2 mM CaCl 2 and 137 mM NaCl at pH 7.0.
  • Waters HPLC systems were used for size exclusion and reversed phase HPLC analyses.
  • 50 ⁇ s of rhASA was injected on to an Agilent Zorbax GF-250 column (4.6 mm ⁇ 250 mm) and run isocratically at 0.24 mL/min using a mobile phase of 100 mM sodium citrate pH 5.5 (octomer detection) with a detection wavelength of 280 nm.
  • the analyses were repeated using mobile phase conditions of 100 mM sodium citrate, pH 7.0 (dimer detection).
  • rhASA Prior to the selection of buffers for long term stability, two “pH memory” experiments were performed to investigate if the protein buffer-exchanged into saline solution was capable of maintaining the pH of the original buffer.
  • rhASA at approximately 8 mg/mL, was first dialyzed into 10 mM citrate-phosphate with 137 mM NaCl, at either a pH value of 5.5 or 7.0, followed by a second dialysis into saline solution.
  • rhASA was dialyzed into 10 mM citrate-phosphate with 137 mM NaCl, at either pH values of 5.5 or 7.0 and subsequently buffer exchanged and concentrated into saline solutions to approximately 35 mg/mL.
  • Polysorbate 20 was included in all five selected solution compositions at a final concentration of 0.005%.
  • the surfactant choice was made based on prior experience of the in vivo tolerability of P20 at 0.005% for CNS delivery of other Shire proteins.
  • a solution of 5% P20 (v/v) was prepared and the appropriate volume was added to each protein formulation to obtain a final concentration of 0.005%.
  • Example preparation As in Table 8. A one year study was initiated in the proposed formulations (Table 9). The stability samples at each time point were analyzed by SEC-HPLC, RP-HPLC, OD320, protein concentration, pH, specific activity, SDS-PAGE (Coomassie), and appearance.
  • Formulation Composition Formulation with 0.005% Polysorbate 20 Study Conditions A 154 mM NaCl, pH 5.9 5° C., 25° C., B 5 mM sodium phosphate, 40° C., and 145 mM NaCl, pH 6.0 frozen baseline C 154 mM NaCl, pH 6.5 at ⁇ 65° C.
  • RhASA formulated in saline, pH 7.0 and 1 mM PBS, pH 7.0 with 2 mM CaCl 2 were also evaluated after 3 months storage at the accelerated condition of 25° C. As shown in FIG. 7 , rhASA undergoes a slight amount of fragmentation in these formulations (with intensity approximately that of the 0.5% BSA impurity spike).
  • Freeze-thaw experiments were conducted by performing three cycles of controlled freeze-thaw, from ambient to ⁇ 50° C. at 0.1° C./min on the shelves of a Vertis Genesis 35EL lyophilizer.
  • One mL aliquots of drug substance formulated at 30 mg/mL in each of the five solution compositions (Table 8) were dispensed into 3 mL glass vials for this study.
  • Drug substance 38 ⁇ 4 mg/mL was used for all freeze-thaw studies.
  • 2 mL aliquots of drug substance were dispensed into 5 mL glass vials with 20 mm Flurotec stoppers.
  • Freeze-thaw stress experiments were conducted either on the shelves of a Virtis Genesis 35EL lyophilizer or on the shelves of a controlled rate freezer (Tenney Jr Upright Test Chamber, Model: TUJR-A-VERV). Three cycles of freezing to ⁇ 50° C. and thawing to 25° C.
  • stoppered glass vials containing the same sample volume were used as a study control.
  • 1 mL aliquots of 1 and 5 mg/mL were dispensed into 2 mL polypropylene tubes and were frozen at ⁇ 65° C. The frozen samples were subsequently thawed on the bench top. The cycle was repeated up to 10 times to mimic any potential stress which may occur with handling of the reference standard aliquots.
  • Drug product agitation studies were conducted in triplicate (in 154 mM NaCl, pH 6.0 with 0.005% P20) and compared with one replicate of drug substance (in 154 mM NaCl, pH 6.0). Shaking studies were also repeated without inclusion of P20 in saline formulation. For these studies, either 1 mL or 3 mL aliquots of drug product at 30 mg/mL were dispensed into 3 mL vials to investigate the effect of shaking as well as the headspace volume on quality of rhASA. For these shaking studies, a speed of 220 rpm was used.
  • drug substance and drug product are not readily susceptible to agitation-induced degradation since it took ⁇ 4 hours of continuous stirring (at setting number 5) and 8 hours of continuous vigorous shaking (at 220 rpm) for a change in appearance to occur.
  • the experiment was performed in triplicate and 5 mM phosphate buffer containing 150 mM sodium chloride, pH 6.0, was titrated side-by-side for comparison. Similarly, drug substance at both concentrations was titrated with 1M sodium hydroxide (NaOH) until a final pH of approximately 6.5 was achieved.
  • drug substance was analyzed by inductively coupling plasma mass spectroscopy (ICP-MS). The buffering capacity of diluted rhASA drug substance was also investigated to ensure that the pH value of solution did not change upon dilution of protein solution. Diluted samples ranging from 30 mg/mL to 1 mg/mL were prepared in 1.5 mL eppendorf tubes and the pH values were measured at the onset of dilution and after one week of storage at 2-8° C.
  • FIG. 10 illustrates the buffering capacity of rhASA drug substance compared to 5 mM sodium phosphate buffer, pH 6.3 with 150 mM sodium chloride when titrated with acid.
  • rhASA is contributing to the buffering capacity of the solution since aspartic acid, glutamic acid, and histidine side chains have the ability to act as proton acceptors and/or donors in order to maintain the solution pH.
  • the buffering capacity of this protein was also previously observed during preformulation studies when the “pH memory” effect was discovered. The retention of pH has been demonstrated several times both at the laboratory scale and at the large scale operations.
  • the buffering capacity for the lower pH values is a direct indication of larger numbers of aspartic acid and glutamic acid residues within a given protein compared to histidine residues. While not wishing to be bound by any theory, this can indeed be the case for arylsulfatase A where there are a total of 45 glutamic as well as aspartic acid residues compared to 18 histidine residues.
  • the buffering capacity of drug substance may also be attributable to residual bound phosphate which was shown to be present in drug substance using ICP-MS.
  • Table 25 demonstrates the amount of residual phosphate present in three different LSDL drug substance lots. This data also confirms the consistency of the ultrafiltration and diafiltration steps for the pilot scale process.
  • FIG. 12 exhibits the observed appearance of diluted rhASA.
  • the 1 mg/mL rhASA solution demonstrated an appearance similar to water while the 30 mg/mL appearance was assessed to be between either Reference Suspensions II and III or III and IV.
  • drug substance was formulated at 38 ⁇ 4 mg/mL in 154 mM NaCl, pH 6.0 and drug product was formulated at 30 ⁇ 3 mg/mL in 154 mM NaCl, pH 6.0 in the presence and absence of 0.005% polysorbate 20.
  • Aliquots of 1 mL of drug substance were dispensed into 5 mL polycarbonate bottles with polypropylene screw closures and stored at ⁇ 65° C., ⁇ 15° C. to ⁇ 25° C., and 2-8° C.
  • This example illustrates repeat dose intrathecal (IT) administration of rhASA from a toxicology and safety pharmacology perspective over a six-month period.
  • the IT test article for this study was rhASA. Thirty-six male and 36 female cynomolgus monkeys were randomly assigned to five treatment groups. The animals in Group 1 were untreated implant device control (port & catheter) and were not dosed with the vehicle or test article; however, these animals were dosed with 0.6 mL of PBS on a schedule matching the test article dosing schedule.
  • test article related changes could be categorized into two major types and were present at all dose levels (1.8, 6.0 and 18.6 mg/dose).
  • this increase was interpreted to be due to the presence of the test article (a protein) in the intrathecal space and in the nervous system tissues. Slight, focal increase of microglial cells in the spinal cord and brain in occasional animals (microgliosis was not observed in any high dose animals).
  • both categories of morphologic changes were interpreted to be a response to the presence of the test article. There was no evidence of neuronal necrosis in any animal. None of the test article related changes were related to any biologically adverse reactions in the brain, spinal cord, spinal nerve roots or ganglia. Specifically, there was no evidence of neuronal necrosis or a biologically important glial response. There were no test article related lesions in the non-nervous system tissues.
  • test article related changes had either entirely resolved or were limited to remnants of the prior increase in the inflammatory response associated with the presence of the test article. There were no adverse morphologic effects in the recovery animals.
  • immunohistochemical staining for Arylsulfatase A (rhASA; the test article) was increased in the brain and spinal cord in various cell types, except neurons, for all test article treated groups at the terminal sacrifice. This increase was also apparent in the Kupffer cells of the liver.
  • the IT test article for this study was rhASA. Thirty-six male and 36 female cynomolgus monkeys were randomly assigned to five treatment groups. The animals in Group 1 were untreated implant device control (port & catheter) and were not dosed with the vehicle or test article; however, these animals were dosed with 0.6 mL of PBS on a schedule matching the test article dosing schedule. The animals in Groups 2-5 received 0.6 mL IT infusion of 0, 3, 10 or 31 mg/mL of rhASA (total dose of 0, 1.8, 6.0, or 18.6 mg) every other week (i.e. a total of 12 doses).
  • the brain was cut in a brain matrix at approximately 3 mm coronal slice thickness.
  • the first slice and every second slice thereafter were fixed in formalin for histopathological evaluation and immunohistochemical analysis.
  • the brain was processed as full coronal sections. These sections included at a minimum the following brain regions.
  • the brain sections are listed in the data tables as sections 1 to 8/9 (a section 9 was provided by the testing facility for some animals). Sectioning varied slightly between animals. The brain sections (1 through 8/9) provided above were the approximate location of the various anatomic areas. The brain sections are listed in the data tables as individual sections, with diagnoses pertinent to that section, to facilitate potential, future additional slide review (if any). During data interpretation, individual brain anatomic sites (as listed above) were compared in order to identify any unique test article effects (i.e. unique to a particular brain region). At TPS, all brain sections from all animals were embedded in paraffin, sectioned at 5 microns, stained with hematoxylin and eosin (H&E) and examined microscopically.
  • H&E hematoxylin and eosin
  • brains from the control and high dose animals were stained with Fluoro-Jade B (a stain increasing the sensitivity of evaluating the brain for neuronal degeneration) and a Bielschowsky's silver stain (a procedure that allows for direct visualization of axons, dendrites and neuronal filaments) and examined.
  • Fluoro-Jade B a stain increasing the sensitivity of evaluating the brain for neuronal degeneration
  • Bielschowsky's silver stain a procedure that allows for direct visualization of axons, dendrites and neuronal filaments
  • the spinal cord (cervical, thoracic and lumber) was cut into one centimeter sections. The first slice and every other slice thereafter were fixed in formalin for histopathological evaluation and immunohistochemical analysis.
  • the spinal cord sections (cervical, thoracic (including the catheter tip) and lumbar) from all animals were sectioned at approximately 5 microns, stained with H&E and examined with transverse and oblique sections taken at each level.
  • Serial spinal cord sections from the control and high dose groups were additionally stained with Bielschowsky's silver stain and anti-GFAP (an immunohistochemical stain that allows for the direct visualization of astrocytes and their processes).
  • Dorsal spinal nerve roots and ganglion were embedded in paraffin, with serial sections stained with H&E.
  • serial sections from the control and high dose groups were stained with Bielschowsky's silver stain.
  • Positive control slides were supplied by the study sponsor.
  • the slides were liver sections from mice injected with rhASA.
  • the positive control slides all showed ample evidence of rhASA in Kupffer cells (sinusoidal macrophages) in the liver.
  • the positive control slides are stored with the other slides from this study. All evaluations of the rhASA stained sections were initially conducted blinded to the treatment group of the animal. This was accomplished by having the pathologist initially read the rhASA stained slides with the animal number on the label obscured (by an assistant with knowledge of the actual animal being evaluated), dictating the score (severity grade) during evaluation, and having the same assistant immediately record the staining score (severity grade) into the data tables.
  • the animal ID was then verified by both the study neuropathologist and the assistant to guarantee accurate data entry. This procedure was conducted so as to not introduce any bias into the judging of the overall intensity of staining with the immunohistochemical stain for the detection of intracellular rhASA.
  • the relative degree of staining of neurons, meningeal macrophages, perivascular macrophages and glial cells (astrocytes and microglial cells but likely predominantly microglial cells) was graded in all the brain and spinal cord sections.
  • the average severity scores at each brain and spinal cord level for each group was totaled (by group) and recorded as a total under the tissue heading Brain, General, rhASA Staining and Spinal Cord, General, rhASA Staining.
  • rhASA staining in neurons of the brain was a measure of the neurons in the cerebral cortex and other nuclear areas in the brain.
  • rhASA staining in meningeal macrophages was evidence of uptake of the test article by meningeal macrophages and/or endogenous rhASA in meningeal macrophages.
  • rhASA staining of perivascular macrophages was a measure of uptake of rhASA by macrophages in the brain/spinal cord (or endogenous rhASA), although it should be noted that the perivascular space in the brain and spinal cord (the Virchow-Robins space) is continuous with the meninges.
  • the grading of rhASA staining in the glial cells was predominantly a measure of uptake of the test article/penetration of the test article into the gray and/or white matter, especially of the cerebral cortex (the corona radiata is the white matter beneath the cerebral cortex).
  • the rhASA staining in the white matter appeared to be in astrocytes and microglial cells.
  • the following grading scheme was used to score the degree of rhASA staining the various cell types (neurons, glial cells, macrophages).
  • rhASA staining levels in the neurons of the brain and spinal cord were the most difficult to specifically define.
  • the rhASA staining demonstrated consistently increased levels of rhASA in the meningeal and brain/spinal cord perivascular macrophages and also within glial cells. There were no detectable differences of rhASA staining in neurons between the control and test article treated animals.
  • test article related changes were either totally resolved or were notably diminished in those animals allowed a one-month period without dosing prior to necropsy.
  • the following microscopic changes were present at an incidence and/or severity that indicated a possible relationship to the test article.
  • test article related changes could be categorized into two major types and were present at all dose levels (1.8, 6.0 and 18.6 mg/dose).
  • test article related changes Slight, focal increase of microglial cells in the spinal cord and brain in occasional animals (microgliosis was not observed in any high dose animals). Both categories of morphologic changes were interpreted to be a response to the presence of the test article. There was no evidence of neuronal necrosis in any animal. None of the test article related changes were related to any biologically adverse reactions in the brain, spinal cord, spinal nerve roots or ganglia. Specifically, there was no evidence of neuronal necrosis or a biologically important glial response. There were no test article related lesions in the non-nervous system tissues. Following a one-month recovery period (a dosing free period), the test article related changes had either entirely resolved or were limited to remnants of the prior increase in the inflammatory response associated with the presence of the test article. There were no adverse morphologic effects in the recovery animals.
  • rhASA Arylsulfatase A
  • This example provides interpretive analysis for serum and CSF concentrations of rhASA and anti-rhASA serum antibodies from Northern Biomedical Research, Inc.
  • the objective of the example was to evaluate repeat dose intrathecal (IT) administration of rhASA from a toxicology and safety pharmacology perspective in juvenile ( ⁇ 12 months of age) cynomolgus monkeys. A total of 12 doses were given in a six month period. Animals were necropsied 24 hours or one-month after the last dose. The study design is shown in Table 30.
  • Quantitation of anti-rhASA antibodies in the serum and CSF from cynomolgus monkeys was conducted using a validated method. Briefly, the assay begins by blocking a MSD streptavidin coated plate, followed by incubation with biotin-labeled rhASA. After a washing step, diluted samples, calibrators, and QCs are added to the plate and incubated. After an additional wash step, SULFO TAG-labelled drug is added and incubated. A final wash step is performed and MSD read buffer is added. Plates are read immediately. The signal data in relative luminescence units (RLU) are analyzed using SOFTMax Pro templates.
  • RLU relative luminescence units
  • rhASA Enzyme-Linked Immunosorbent Assay
  • This enzyme-substrate reaction is stopped by the addition of 2N sulfuric acid (H 2 SO4) and the absorbance of each well is measured at the absorbance wavelength 450 nm with a reference wavelength 655 nm.
  • concentrations of ASA in samples are calculated using the rhASA calibration curve in the same plate.
  • the quantitation limit for rhASA in cynomolgus monkey serum is 39.1 ng/mL, and all serum samples from Groups 1 and 2 were below quantitation limit (BQL), see Table 33.
  • Serum levels of rhASA were tested prior to and at 24 hours after Doses 2, 4, 6, 8, 10, and 12 (6-month necropsy), midway through the recovery period, and prior to the recovery necropsy.
  • rhASA levels were undetectable in Group 3 (1.8 mg/dose), Group 4 (6.0 mg/dose), and Group 5 (18.6 mg/dose) prior to Doses 2, 4, 6, 8, 10, and 12, After Dose 12, midway through the recovery period, and prior to the recovery necropsy.
  • rhASA levels were undetectable. Serum levels of rhASA declined in Group 4 (6.0 mg/dose) after Dose 4 and in Group 5 (18.6 mg/dose) after Doses 4 and 6 for males and Doses 4, 6, 8, and 10 for females. This apparent decline in serum rhASA levels may be related to the increasing concentration of anti-rhASA antibodies. There were no apparent sex differences in serum levels of rhASA, given the sample variability and small group numbers in this study.
  • the quantitation limit for rhASA in cynomolgus monkey CSF is 19.5 ng/mL, and all CSF samples from Groups 1 and 2 were BQL, see Table 34.
  • rhASA was detectable in CSF prior to and after Doses 2, 4, 6, 8, 10, and 12 (6-month necropsy) in all dosed groups. The levels were higher postdose (approximately 24 hours postdose) and were dose related. The levels in CSF were much greater than those in serum. There were no apparent sex differences in CSF levels of rhASA, given the sample variability and small group numbers in this study. rhASA was not detectable midway through the recovery period and prior to the recovery necropsy in all dosed groups.
  • CSF levels at the Dose 12 (necropsy) collections for rhASA treated groups were lower than levels postdose 8 and 11.
  • Potential reasons for lower rhASA levels at necropsy include the larger volume taken ( ⁇ 2.25 mL total for cell counts, chemistry, rhASA and anti-rhASA antibody) at necropsy vs. those taken at in-life dosing interval (up to 0.5 mL pre- or postdose for rhASA concentration). Additionally, some animals did not have patent catheters at necropsy, and samples were taken via a CM tap rather than via the catheter. This route consistently yielded lower rhASA concentrations as compared with sampling via the catheter.
  • Anti-rhASA antibodies in serum were detected in every animal treated with rhASA at some time point, see Table 35. Animals are defined as positive for anti-rhASA antibodies if the level of anti-rhASA antibody was above the quantitation limit (78.1 ng/mL). Animals remained positive for anti-rhASA antibodies once they seroconverted. No animals were positive for anti-rhASA antibodies at the predose 2 timepoint. All rhASA animals except Male No. 026 (Group 4; 6.0 mg/dose) were positive for serum anti-rhASA antibodies at the predose 4 timepoint. Male No. 026 was positive for serum antiboday at the predose 6 timepoint. In Group 5 (18.6 mg/kg), the necropsy antibody samples had lower antibody levels.
  • This apparent decrease may be due to the presence of rhASA interfering with the assay.
  • the titer was generally higher in the mid- and high-dose groups (6.0 and 18.6 mg/dose) than the low dose animals (1.8 mg/dose).
  • the presence of anti-rhASA antibodies is an expected result from treating cynomolgus monkeys with a recombinant human protein′. Given the variability in the results, there was no apparent sex differences.
  • mice 11 Wild-type control (mASA+/+hASA ⁇ / ⁇ ) mice were assigned to Group A and received no treatment.
  • thirty-four (34) hASAC69S/ASA ⁇ / ⁇ mice were assigned to each of 5 dose groups and received vehicle (Group B) or rhASA (rhASA) at doses of 20 mg/kg (intravenous [IV]; Group C) or 0.04, 0.12, and 0.21 mg (Groups D, E, and F, respectively) on Days 1, 9, 15/16, and 22. All IV doses were administered via a tail vein. All intrathecal (IT) doses were administered as an infusion in a volume of 12 ⁇ L at an approximate range of 2 ⁇ L/20 seconds (Table 40).
  • the ASA knockout mouse hASAC69S/ASA( ⁇ / ⁇ ) is an accepted model of MLD, and has been used to test potential treatments for this disease.
  • the intrathecal route is the intended route of administration in humans.
  • the intravenous route of administration has been tested for this compound and a similar compound in MLD mice.
  • An intravenous control group has been added as a positive control for histological changes expected in peripheral organs.
  • Animals received 100, 300, or 520 mg/kg of brain weight (0.04, 0.12, 0.21 mg, respectively) of rhASA.
  • the dose levels normalized to brain weight selected for this study correspond to doses that are planned for use in humans or have been used in toxicology studies or in previous efficacy models of lysosomal storage diseases. These doses were not expected to have any toxicity.
  • mice Mus musculus .
  • Strain hASAC69S/ASA ( ⁇ / ⁇ ) mice and wild type controls Age Approximately 14-17 months at arrival No. of Groups 6 No. of Animals 34 ASA knockout mice + 11 wild type controls Following arrival, each animal was examined to assess health status. Housing Animals were group housed in high-temp polycarbonate filter-top cages, with CareFresh paper bedding and water bottles. Each cage was clearly labeled with a cage card indicating project, group and animal numbers, and sex. Each animal was uniquely identified using an ear punch system. Animals were treated in compliance with federal guidelines. The targeted conditions for animal room environment and photoperiod were as follows:
  • the photoperiod may have been temporarily interrupted for scheduled activities. Such interruptions are not considered to affect the outcome or quality of this research.
  • ASA ( ⁇ / ⁇ ) hASA (+/ ⁇ ) animals were assigned consecutive numbers (1 through 34) as they were removed from their cages, weighed, and ear punched during acclimation. Animals were then assigned to the treatment groups using Research Randomizer (www.randomizer.org) on Jan. 3, 2011. the first 9 numbers were assigned to Group B, the next 5 to Group C, the next 5 to Group D, the next 5 to Group E, and the final 10 to Group F. Animals were assigned as follows in Table 41:
  • Test Article Identity rhASA Description human recombinant Arylsulfatase A (rhASA) Storage Conditions Approximately 4° C. Vehicle Identity rhASA Vehicle (154 mM NaCl, 0.005% polysorbate 20, pH ⁇ 6.0) Storage Condition Approximately 4° C. Preparation of Vehicle
  • the vehicle was used as provided. The vehicle was warmed on the bench top (ambient). Once the vehicle was warmed, the material was mixed by gently swirling and inverting. The bottles were not vortexed or shaken. The bottle was dried before accessing the material. Any remaining vehicle was returned to the refrigerator (1° C.-8° C.).
  • rhASA was diluted with vehicle to achieve the necessary concentrations.
  • the test article was warmed on the bench top (ambient). Once the test article was warmed, the material was mixed by gently swirling and inverting. The bottles were not vortexed or shaken.
  • An infrared dye (such as IRDye®, LI-COR Biosciences, Lincoln, Nebr.) was utilized for tracking the injections. Dyes such as this have been used in intrathecal injections as a survival procedure after intrathecal administration. The dye was mixed with the test article before administration; 1 nmole of dye in 1 ⁇ L was added to the test article. In addition to the infrared dye, 1 ⁇ L of FD&C blue #1 (0.25%) was used for tracking injections. This blue dye is a common food additive and is generally considered safe and non-toxic.
  • mice were anesthetized using 1.25% 2,2,2 tribromoethanol (Avertin) at 200-300 ⁇ L/10 grams body weight (250-350 mg/kg) by intraperitoneal injection.
  • Dorsal hair was removed between the base of the tail and the shoulder blades using a clippers.
  • the shaved area was cleaned with povidine/betadine scrub followed by isopropyl alcohol.
  • a small midline skin incision (1-2 cm) was made over the lumbosacral spine, and the intersection of the dorsal midline and the cranial aspect of the wings of the ilea (singular ileum) was identified.
  • the muscle in the iliac fossa is a heart shaped muscle.
  • mice were anesthetized using isoflurane, if required, and were placed in a restrainer.
  • the tail vein was dilated by warming by flicking the tail gently with the finger.
  • the injection site was then wiped with 70% ethanol.
  • the animal was placed in a warm chamber (40° C.) for 1-1.5 minutes.
  • a 28- to 30-gauge needle was used to inject test material.
  • the volume of injection was 5-10 mL/kg.
  • mice in Groups B-F were euthanized. Animals were subjected to different tissue collection procedures, as detailed below. Animals in Group A were not treated; however, they were euthanized on Jan. 27 or 28, 2011 and subjected to tissue collection procedures, as detailed below.
  • a terminal blood sample (approximately 0.5 mL) was collected from all animals (Groups A-F) via retroorbital puncture under isoflurane anesthesia.
  • a glass tube was placed in the orbit, gently penetrating the area behind the eye and thus disrupting the venous drainage located behind the eye. Blood was collected by capillary action and/or gravity flow. Following blood collection, pressure was applied to the orbit to stop the bleeding.
  • the whole blood samples were processed to serum and frozen at ⁇ 80° C.
  • the serum was stored at ⁇ 80° C. and analyzed for antibodies.
  • mice were transcardially perfused with heparinized saline solution (1 U/mL sodium heparin in 0.9% NaCl, sterile-filtered) chilled ice-cold and then with 4% paraformaldehyde at approximately 4° C.
  • the brain was removed, and the abdomen was cut to expose the internal organs further. The brain and carcass were placed in paraformaldehyde, except for the tail snip which was frozen.
  • Tissues for Lipid Analysis (Groups A, B, and F; 6, 4, and 5 Animals, Respectively)
  • mice were transcardially perfused with heparinized saline solution (1 U/mL sodium heparin in 0.9% NaCl, sterile-filtered) chilled ice-cold. Exemplary tissues collected for lipid analyses are presented in Table 42.
  • rhASA reduced sulfatide storage in the spinal cord of MLD mice, particularly in the white matter, FIG. 19 .
  • Morphometry analysis of the spinal cord demonstrated that the optical density of alcian blue staining was statistically significantly reduced after rhASA dosing, FIG. 20 .
  • rhASA treated MLD mice also exhibited reduced lysosomal activity in the brain, FIG. 21 . This reduction was statistically significant in the high-dose group (0.21 mg-520 mg/kg brain weight) compared with vehicle treated animals, FIG. 22 .
  • Immunotolerant MLD mice hASAC69S/ASA( ⁇ / ⁇ )
  • rhASA rhASA one time each week for 4 weeks (a total of 4 doses).
  • Doses were vehicle (154 mM NaCl, 0.005% polysorbate 20, pH ⁇ 6.0), 0.04, 0.12, 0.21 mg/dose (normalized doses were 100, 300 and 520 mg/kg of brain weight, respectively).
  • At terminal timepoints efficacy was evaluated by immunohistochemistry assessment of sulfatide clearance and lysosome activity within the brain and spinal cord.
  • lysosomal-associated membrane protein an indicator of lysosomal processes. Additionally, morphometry analysis was performed on alcian blue and LAMP stained sections of the spinal cord (cervical, thoracic and lumbar) and brain.
  • rhASA treated MLD mice exhibit evidence of improvement within the histological markers of disease, such as reduced sulfatide storage (noted by alcian blue staining) and lysosomal activity in the brain. These histopathological changes were observed near the site of administration (spinal cord) as well as in the distal portions of the brain.
  • the brains were cut in a brain matrix at 3 mm thick coronal slice thickness. Each brain was sectioned into full coronal slices including: neocortex (including frontal, parietal, temporal, and occipital cortex), paleocortex (olfactory bulbs and/or piriform lobe), basal ganglia (including caudate and putamen), limbic system (including hippocampus and cingulate gyri), thalamus/hypothalamus, midbrain regions (including substantia nigra), cerebellum, pons, and medulla oblongata.
  • the locations from which individual tissue samples were obtained (via 4-mm biopsy punch) are shown in FIGS. 32-37 .
  • FIGS. 32-37 are from the University of Wisconsin and Michigan State Comparative Mammalian Brain Collections, (also the National Museum of Health and Medicine). Punch number 22 was not collected, as this structure was not present during necropsy. All brain samples were frozen and stored at ⁇ 60° C. or below prior to analysis for rhASA using an enzyme-linked immunosorbent assay.
  • the first brain slice and every second slice thereafter were fixed in formalin for histopathological evaluation and immunohistochemical.
  • the second brain slice and every second slice thereafter were frozen for test article concentration analysis.
  • samples of brain Prior to freezing, samples of brain were taken from the right portion of the even-numbered, test article analysis brain slices for biodistribution analysis. The location of the brain samples were photographed at necropsy and the brain slice number was recorded. The samples were obtained using either a 4-mm circular punch or cut with a scalpel to optimize the amount of white matter collected. All punches were frozen and stored at ⁇ 60° C. or below for test article analysis. The remainder of the brain slice was frozen and stored at ⁇ 60° C. or below for possible test article analysis. Locations of the punches are shown in Appendix B.
  • the spinal cord (cervical, thoracic and lumbar) was cut into one-centimeter sections. The first slice and every second slice thereafter was fixed in formalin for histopathological and immunohistochemical analysis. The second slice of spinal cord and every second slice thereafter was frozen and stored at ⁇ 60° C. or lower for test article analysis. The distribution of slices was adjusted so that the slice with the tip of the intrathecal catheter (Slice 0) was fixed in formalin and analyzed for histopathology.
  • Tissue samples were homogenized in lysis buffer, centrifuged to remove any tissue debris, and stored at ⁇ 80° C. until assayed.
  • rhASA concentration in the soluble fractions of the homogenates was determined by an ELISA using polyclonal rabbit antibody SH040 as the capture antibody and HRP (horseradish peroxidase)-conjugated anti-ASA monoclonal antibody 19-16-3 as the detection antibody.
  • TMB tetramethylbenzidine
  • Samples were also analyzed by a bicinchoninic acid (BCA) protein determination assay to obtain the concentration of protein in an unknown sample.
  • BCA bicinchoninic acid
  • the protein concentration for each sample was determined by interpolation of an albumin standard curve. rhASA concentration results were then normalized to total protein in tissue extracts, as determined by bicinchoninic acid assay.
  • the ASA levels of all punches for the vehicle, 1.8 mg/dose, 6.0 mg/dose, and 18.6 mg/dose groups are shown in FIG. 23 , FIG. 24 , FIG. 25 , and FIG. 26 , respectively.
  • the ASA levels of all punches for the recovery animals for the device control, vehicle, 1.8 mg/dose, 6.0 mg/dose, and 18.6 mg/dose groups are shown in FIG. 27 , FIG. 28 , FIG. 29 , FIG. 30 , and FIG. 31 , respectively
  • ASA levels for selected punches that were taken near the surface (meninges) of the brain are shown in FIG. 32 .
  • ASA levels for selected punches that are considered to contain mostly deep white brain matter are shown in FIG. 33 .
  • White matter is composed of bundles of myelinated nerve cell processes (or axons).
  • Selected punches which contain mostly material from the deep grey brain matter are shown in FIG. 34 .
  • Grey matter contains neural cell bodies, in contrast to white matter.
  • the values of ASA in selected punches from the surface, deep white and deep grey are shown for each dose group in FIG. 35 .
  • Liver concentration data is shown in FIG. 37 .
  • ASA concentration levels in the liver, spinal cord, and brain of the device and vehicle-dosed control groups were in some cases measurable.
  • the levels in liver and spinal cord were lower than any of the rhASA-treated groups ( FIG. 23 , FIG. 32 , and FIG. 33 ).
  • the level of rhASA measured in the device control and vehicle-dosed animals represents a cross-reactivity between the anti-rhASA antibody used in the ELISA with the native cynomolgus monkey protein.
  • the reported values in the device control and vehicle tissues do not represent quantitative values for cynomolgus monkey rhASA in the tissues, because the degree of cross-reactivity between the antibody and cynomolgus ASA is not known, and the fact that the assay standards use human ASA.
  • the variation in the levels of ASA detected between device control and vehicle-dosed tissues may be interpreted as demonstrated variability in the relative amounts of cynomolgus ASA in different tissues and anatomical regions.
  • the ASA levels in spinal cord slices ranged from 160-2352, 1081-6607, and 1893-9252 ng/mg protein in males and 0-3151, 669-6637, and 1404-16424 ng/mg protein in females for the 1.8, 6.0, and 18.6 mg/dose groups, respectively ( FIG. 32 ).
  • Levels of ASA were higher in the lumbar region of the spine than in the cervical region.
  • Levels of ASA protein detected in the liver were dose responsive in the rhASA treated groups and were very low in the vehicle group.
  • Mean ASA levels were 88, 674, and 2424 in males and 140, 462, and 1996 ng/mg protein in females for the 1.8, 6.0, and 18.6 mg/dose groups, respectively ( FIG. 33 ).
  • ASA level of ASA appeared to be dose-related in samples prepared from the spinal cord slices and liver of the rhASA-dosed groups. Many of the brain regions tested demonstrated a clear dose relationship between ASA levels and rhASA administration, while others were more equivocal. In general, ASA levels in the brain increased with rhASA dose.
  • the objective this study is to evaluate the pharmacokinetic (PK) and biodistribution of various therapeutic replacement enzymes after intrathecal (IT) and intravenous (IV) administration to cynomolgus monkeys.
  • Exposure to ASA in serum increases in a more than proportional manner following IT-L administration.
  • CSF SERUM PARTITION CSF: PLASMA PARTITION ARYLSULFATASE A ARYLSULFATASE A ARYLSULFATASE A ARYLSULFATASE A (PHASE 1B: IV (PHASE 1B: IT-L (PHASE 1B: IT-L (PHASE 1B: IT-L 1 MG/KG) 1.8 MG) 6 MG) 18.6 MG) 0.74 NC 445 452
  • Direct CNS administration through, e.g., IT delivery can be used to effectively treat MLD patients.
  • This example illustrates a multicenter dose escalation study designed to evaluate the safety of up to 3 dose levels every other week (EOW) for a total of 40 weeks of rhASA administered via an intrathecal drug delivery device (IDDD) to patients with late infantile MLD.
  • IDDD intrathecal drug delivery device
  • FIGS. 45-48 Various exemplary intrathecal drug delivery devices suitable for human treatment are depicted in FIGS. 45-48 .
  • Patients are selected for the study based on inclusion of the following criteria: (1) appearance of first symptoms prior to 30 months of age; (2) ambulatory at the time of screening (defined as the ability to stand up alone and walk forward 10 steps with one hand held); (3) presence of neurological signs at time of screening. Typically, patients' history of hematopoietic stem cell transplantation are excluded.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Molecular Biology (AREA)
  • Psychology (AREA)
  • Inorganic Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Hospice & Palliative Care (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Psychiatry (AREA)
  • Obesity (AREA)
  • Dermatology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
US13/168,963 2010-06-25 2011-06-25 Methods and compositions for CNS delivery of arylsulfatase A Active 2031-11-30 US9770410B2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/168,963 US9770410B2 (en) 2010-06-25 2011-06-25 Methods and compositions for CNS delivery of arylsulfatase A
US15/684,568 US10646554B2 (en) 2010-06-25 2017-08-23 Methods and compositions for CNS delivery of arylsulfatase A
US16/828,731 US20200405825A1 (en) 2010-06-25 2020-03-24 Methods and Compositions for CNS Delivery of Arylsulfatase A

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US35885710P 2010-06-25 2010-06-25
US36078610P 2010-07-01 2010-07-01
US38786210P 2010-09-29 2010-09-29
US201161435710P 2011-01-24 2011-01-24
US201161442115P 2011-02-11 2011-02-11
US201161476210P 2011-04-15 2011-04-15
US201161495268P 2011-06-09 2011-06-09
US13/168,963 US9770410B2 (en) 2010-06-25 2011-06-25 Methods and compositions for CNS delivery of arylsulfatase A
PCT/US2011/041926 WO2011163650A2 (en) 2010-06-25 2011-06-25 Methods and compositions for cns delivery of arylsulfatase a

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/684,568 Continuation US10646554B2 (en) 2010-06-25 2017-08-23 Methods and compositions for CNS delivery of arylsulfatase A

Publications (2)

Publication Number Publication Date
US20120009171A1 US20120009171A1 (en) 2012-01-12
US9770410B2 true US9770410B2 (en) 2017-09-26

Family

ID=46888844

Family Applications (1)

Application Number Title Priority Date Filing Date
US13/168,963 Active 2031-11-30 US9770410B2 (en) 2010-06-25 2011-06-25 Methods and compositions for CNS delivery of arylsulfatase A

Country Status (25)

Country Link
US (1) US9770410B2 (zh)
EP (3) EP3626257B1 (zh)
JP (1) JP6045493B2 (zh)
KR (5) KR102537084B1 (zh)
CN (1) CN103282046B (zh)
AU (4) AU2011270670B2 (zh)
BR (1) BR112012033216B1 (zh)
CA (1) CA2803003C (zh)
CY (2) CY1122282T1 (zh)
DK (1) DK2585104T3 (zh)
ES (1) ES2754776T3 (zh)
HR (1) HRP20191923T1 (zh)
IL (3) IL291556B2 (zh)
LT (1) LT2585104T (zh)
ME (1) ME03647B (zh)
MX (1) MX344795B (zh)
NZ (2) NZ702799A (zh)
PL (2) PL3626257T3 (zh)
RS (1) RS59468B1 (zh)
RU (1) RU2671503C2 (zh)
SI (1) SI2585104T1 (zh)
TW (1) TWI667037B (zh)
UA (1) UA115650C2 (zh)
WO (1) WO2011163650A2 (zh)
ZA (1) ZA201300675B (zh)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180036388A1 (en) * 2013-05-15 2018-02-08 Regenxbio Inc. Adeno-associated virus mediated gene transfer to the central nervous system
WO2021061517A1 (en) * 2019-09-23 2021-04-01 University Of Washington Assay of arylsulfatase a enzymatic activity in dried blood spots by mass spectrometry and fluorometry
US11020461B2 (en) * 2016-02-17 2021-06-01 Takeda Pharmaceutical Company Limited Methods and compositions for CNS delivery of arylsulfatase A
US11471516B2 (en) * 2010-06-25 2022-10-18 Shire Human Genetic Therapies, Inc. CNS delivery of therapeutic agents
US11819539B2 (en) 2017-09-22 2023-11-21 The Trustees Of The University Of Pennsylvania Gene therapy for treating Mucopolysaccharidosis type II

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20140005842A (ko) 2010-06-25 2014-01-15 샤이어 휴먼 지네틱 테라피즈 인크. 헤파란 n-설파타제의 cns 전달을 위한 방법들 및 조성물들
ES2650689T3 (es) 2010-06-25 2018-01-19 Shire Human Genetic Therapies, Inc. Administración de agentes terapéuticos al sistema nervioso central
EP3626257B1 (en) 2010-06-25 2021-08-04 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of arylsulfatase a
EP3964229A1 (en) 2010-06-25 2022-03-09 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of iduronate-2-sulfatase
US20140377244A1 (en) 2011-12-23 2014-12-25 Shire Human Genetic Therapies, Inc. Stable formulations for cns delivery of arylsulfatase a
EP2943568B1 (en) * 2013-01-09 2019-11-20 Shire Human Genetic Therapies, Inc. Methods for purification of arylsulfatase a
US10208734B2 (en) * 2015-04-23 2019-02-19 Continuum Dynamics, Inc. Lift-driven wind turbine with force canceling blade configuration
AU2016292759B2 (en) * 2015-07-10 2021-05-20 Byondis B.V. Compositions comprising antibody-duocarmycin drug conjugates
KR102473887B1 (ko) * 2016-06-01 2022-12-06 세르비에 아이피 유케이 리미티드 폴리알킬렌 옥시드-아스파라기나제의 제형, 및 그의 제조 및 사용 방법
CA3048381A1 (en) * 2016-12-28 2018-07-05 Jcr Pharmaceuticals Co., Ltd. Lyophilized preparation
CN111699249A (zh) 2017-12-19 2020-09-22 夏尔人类遗传性治疗公司 纯化的芳基硫酸酯酶a及其组合物
CN112566652A (zh) 2018-06-25 2021-03-26 Jcr制药股份有限公司 含有蛋白的水性液体制剂

Citations (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4743265A (en) 1986-04-23 1988-05-10 Dij Catheter Corp Articulated catheter placement device
US5972333A (en) 1995-04-19 1999-10-26 Women's And Children Hospital Synthetic mammalian sulphamidase and genetic sequences encoding same
US6118045A (en) 1995-08-02 2000-09-12 Pharming B.V. Lysosomal proteins produced in the milk of transgenic animals
US6217552B1 (en) 1999-03-01 2001-04-17 Coaxia, Inc. Medical device for selective intrathecal spinal cooling in aortic surgery and spinal trauma
US20020052311A1 (en) 1999-09-03 2002-05-02 Beka Solomon Methods and compostions for the treatment and/or diagnosis of neurological diseases and disorders
US20020099025A1 (en) 1999-12-30 2002-07-25 Heywood James A. Treatment of neurological disorders
WO2002087510A2 (en) 2001-04-30 2002-11-07 Symbiontics, Inc. Subcellular targeting of therapeutic proteins
US6534300B1 (en) 1999-09-14 2003-03-18 Genzyme Glycobiology Research Institute, Inc. Methods for producing highly phosphorylated lysosomal hydrolases
US20030072761A1 (en) 2001-10-16 2003-04-17 Lebowitz Jonathan Methods and compositions for targeting proteins across the blood brain barrier
WO2003032913A2 (en) 2001-10-16 2003-04-24 Symbiontics Inc. Methods and compositions for targeting proteins across the blood brain barrier
RU2209080C2 (ru) 1997-08-22 2003-07-27 Сейкагаку Корпорейшн Фармацевтическая композиция, включающая фермент, разрушающий гликозаминогликаны, и применение указанного фермента для получения лекарственного препарата для терапии грыжи межпозвоночного диска
WO2003102583A1 (en) 2002-05-29 2003-12-11 Symbiontics, Inc. Targeted therapeutic proteins
US20040006008A1 (en) 2001-04-30 2004-01-08 Symbiontics, Inc. Targeted therapeutic proteins
US20040005309A1 (en) 2002-05-29 2004-01-08 Symbiontics, Inc. Targeted therapeutic proteins
WO2004043373A2 (en) 2002-11-07 2004-05-27 Biomarin Pharmaceutical Inc. Precursor n-acetylgalactosamine-4-sulfatase, methods of treatment using said enzyme and methods for producing and purifying said enzyme
US20040248262A1 (en) 2003-01-22 2004-12-09 Koeberl Dwight D. Constructs for expressing lysomal polypeptides
WO2005002515A2 (en) 2003-06-20 2005-01-13 Biomarin Pharmaceutical Inc. Delivery of therapeutic compounds to the brain and other tissues
US20050042227A1 (en) 2003-06-20 2005-02-24 Todd Zankel Megalin-based delivery of therapeutic compounds to the brain and other tissues
US20050048047A1 (en) * 2003-08-29 2005-03-03 Kakkis Emil D. Delivery of therapeutic compounds to the brain and other tissues
WO2005078077A2 (en) 2004-02-10 2005-08-25 Zystor Therapeutics, Inc. Acid alpha-glucosidase and fragments thereof
US20050208090A1 (en) 2004-03-18 2005-09-22 Medtronic, Inc. Methods and systems for treatment of neurological diseases of the central nervous system
US20050281805A1 (en) 2002-05-29 2005-12-22 Symbiontics, Inc. Targeted therapeutic proteins
US20060029656A1 (en) 2004-02-03 2006-02-09 Biodelivery Sciences International, Inc. Replacement enzyme cochleates
US20060177433A1 (en) 2002-04-25 2006-08-10 Treco Douglas A Treatment of alpha-galactosidase a deficiency
WO2007141346A2 (en) 2006-06-09 2007-12-13 Novartis Ag Genes regulating intracellular cholesterol trafficking as targets for treatment of cholesterol-related diseases
US20080003211A1 (en) 2004-01-30 2008-01-03 Zymenex A/S Production and Purification of Recombinant Arylsulftase
US7351410B2 (en) 1995-08-02 2008-04-01 Genzyme Therapeutic Products Limited Partnership Treatment of Pompe's disease
WO2008070769A1 (en) 2006-12-06 2008-06-12 Medtronic, Inc. Intrathecal catheter
US20090041741A1 (en) 2007-03-06 2009-02-12 Saint Louis University Modified enzyme and treatment method
US20090130079A1 (en) 2006-01-20 2009-05-21 Genzyme Corporation Intraventricular enzyme delivery for lysosomal storage diseases
US20090191178A1 (en) 2004-02-06 2009-07-30 Biomarin Pharmaceutical Inc. Manufacture of Highly Phosphorylated Lysosomal Enzymes and Uses Thereof
US20090246187A1 (en) 2006-04-04 2009-10-01 Shire Pharmaceuticals Ireland Limited Process for concentration of a polypeptide
WO2009137721A2 (en) 2008-05-07 2009-11-12 Zystor Therapeutics, Inc. Lysosomal targeting peptides and uses thereof
US20090291062A1 (en) 2007-11-30 2009-11-26 Wolfgang Fraunhofer Protein formulations and methods of making same
US20090297592A1 (en) 2005-05-11 2009-12-03 Jcr Pharmaceuticals Co., Ltd. Lipid Vesicle Composition
US20100068195A1 (en) 2008-01-18 2010-03-18 Biomarin Pharmaceutical Inc. Manufacture of Active Highly Phosphorylated Human Lysosomal Sulfatase Enzymes and Uses Thereof
US20100260706A1 (en) 2007-08-15 2010-10-14 Oren Bogin Compositions and methods for improving production of recombinant polypeptides
US20110105560A1 (en) 2005-06-08 2011-05-05 Amicus Therapeutics, Inc. Treatment of cns disorders associated with mutations in genes encoding lysosomal enzymes
WO2011163649A2 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc Methods and compositions for cns delivery of iduronate-2-sulfatase
US20110318324A1 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of b-galactocerebrosidase
US20110318327A1 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Treatment of sanfilippo syndrome type b
WO2011163650A2 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of arylsulfatase a
WO2011163648A1 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Cns delivery of therapeutic agents
WO2011163647A2 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of heparan n-sulfatase
WO2012023623A2 (ja) 2010-08-19 2012-02-23 日本ケミカルリサーチ株式会社 ハンター症候群の治療剤
US20130168961A1 (en) 2009-06-29 2013-07-04 LighSail Energy Inc. Compressed air energy storage system utilizing two-phase flow to facilitate heat exchange
US20130295077A1 (en) 2010-06-25 2013-11-07 Shire Human Genetic Therapies, Inc. Treatment of sanfilippo syndrome type b

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101410408A (zh) * 2006-04-04 2009-04-15 希尔制药爱尔兰有限责任公司 用于浓缩多肽的方法

Patent Citations (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4743265A (en) 1986-04-23 1988-05-10 Dij Catheter Corp Articulated catheter placement device
US5972333A (en) 1995-04-19 1999-10-26 Women's And Children Hospital Synthetic mammalian sulphamidase and genetic sequences encoding same
US6118045A (en) 1995-08-02 2000-09-12 Pharming B.V. Lysosomal proteins produced in the milk of transgenic animals
US7351410B2 (en) 1995-08-02 2008-04-01 Genzyme Therapeutic Products Limited Partnership Treatment of Pompe's disease
US20040172665A1 (en) 1995-08-02 2004-09-02 Reuser Arnold J.J. Compositions and methods for treating enzyme deficiency
RU2209080C2 (ru) 1997-08-22 2003-07-27 Сейкагаку Корпорейшн Фармацевтическая композиция, включающая фермент, разрушающий гликозаминогликаны, и применение указанного фермента для получения лекарственного препарата для терапии грыжи межпозвоночного диска
US6217552B1 (en) 1999-03-01 2001-04-17 Coaxia, Inc. Medical device for selective intrathecal spinal cooling in aortic surgery and spinal trauma
US20040243058A1 (en) 1999-03-01 2004-12-02 Coaxia, Inc. Medical device for selective intrathecal spinal cooling in aortic surgery and spinal trauma
US20020052311A1 (en) 1999-09-03 2002-05-02 Beka Solomon Methods and compostions for the treatment and/or diagnosis of neurological diseases and disorders
US6534300B1 (en) 1999-09-14 2003-03-18 Genzyme Glycobiology Research Institute, Inc. Methods for producing highly phosphorylated lysosomal hydrolases
US6537785B1 (en) 1999-09-14 2003-03-25 Genzyme Glycobiology Research Institute, Inc. Methods of treating lysosomal storage diseases
US20020099025A1 (en) 1999-12-30 2002-07-25 Heywood James A. Treatment of neurological disorders
US20040006008A1 (en) 2001-04-30 2004-01-08 Symbiontics, Inc. Targeted therapeutic proteins
US20030082176A1 (en) 2001-04-30 2003-05-01 Symbiontics, Inc. Subcellular targeting of therapeutic proteins
US7560424B2 (en) 2001-04-30 2009-07-14 Zystor Therapeutics, Inc. Targeted therapeutic proteins
US7396811B2 (en) 2001-04-30 2008-07-08 Zystor Therapeutics, Inc. Subcellular targeting of therapeutic proteins
WO2002087510A2 (en) 2001-04-30 2002-11-07 Symbiontics, Inc. Subcellular targeting of therapeutic proteins
WO2003032727A1 (en) 2001-10-16 2003-04-24 Symbiontics Inc. Methods and compositions for targeting underglycosylated proteins across the blood brain barrier
WO2003032913A2 (en) 2001-10-16 2003-04-24 Symbiontics Inc. Methods and compositions for targeting proteins across the blood brain barrier
US20030072761A1 (en) 2001-10-16 2003-04-17 Lebowitz Jonathan Methods and compositions for targeting proteins across the blood brain barrier
US20060177433A1 (en) 2002-04-25 2006-08-10 Treco Douglas A Treatment of alpha-galactosidase a deficiency
US20040005309A1 (en) 2002-05-29 2004-01-08 Symbiontics, Inc. Targeted therapeutic proteins
US20050281805A1 (en) 2002-05-29 2005-12-22 Symbiontics, Inc. Targeted therapeutic proteins
US7629309B2 (en) 2002-05-29 2009-12-08 Zystor Therapeutics, Inc. Targeted therapeutic proteins
WO2003102583A1 (en) 2002-05-29 2003-12-11 Symbiontics, Inc. Targeted therapeutic proteins
WO2004043373A2 (en) 2002-11-07 2004-05-27 Biomarin Pharmaceutical Inc. Precursor n-acetylgalactosamine-4-sulfatase, methods of treatment using said enzyme and methods for producing and purifying said enzyme
US20040248262A1 (en) 2003-01-22 2004-12-09 Koeberl Dwight D. Constructs for expressing lysomal polypeptides
US20050042227A1 (en) 2003-06-20 2005-02-24 Todd Zankel Megalin-based delivery of therapeutic compounds to the brain and other tissues
WO2005002515A2 (en) 2003-06-20 2005-01-13 Biomarin Pharmaceutical Inc. Delivery of therapeutic compounds to the brain and other tissues
US7442372B2 (en) 2003-08-29 2008-10-28 Biomarin Pharmaceutical Inc. Delivery of therapeutic compounds to the brain and other tissues
US20050048047A1 (en) * 2003-08-29 2005-03-03 Kakkis Emil D. Delivery of therapeutic compounds to the brain and other tissues
WO2005021064A2 (en) 2003-08-29 2005-03-10 Biomarin Pharmaceutical Inc. Delivery of therapeutic compounds to the brain and other tissues
US20120148558A1 (en) 2003-08-29 2012-06-14 Biomarin Pharmaceutical Inc. Delivery of Therapeutic Compounds to the Brain and Other Tissues
US20090017005A1 (en) 2003-08-29 2009-01-15 Biomarion Pharmaceutical Inc. Delivery of Therapeutic Compounds to the Brain and Other Tissues
US20080003211A1 (en) 2004-01-30 2008-01-03 Zymenex A/S Production and Purification of Recombinant Arylsulftase
US20060029656A1 (en) 2004-02-03 2006-02-09 Biodelivery Sciences International, Inc. Replacement enzyme cochleates
US20090191178A1 (en) 2004-02-06 2009-07-30 Biomarin Pharmaceutical Inc. Manufacture of Highly Phosphorylated Lysosomal Enzymes and Uses Thereof
WO2005078077A2 (en) 2004-02-10 2005-08-25 Zystor Therapeutics, Inc. Acid alpha-glucosidase and fragments thereof
US20080299640A1 (en) 2004-02-10 2008-12-04 Zystor Therapeutics, Inc. Acid alpha-glucosidase and fragments thereof
US20050244400A1 (en) 2004-02-10 2005-11-03 Zystor Therapeutics, Inc. Acid alpha-glucosidase and fragments thereof
US20050208090A1 (en) 2004-03-18 2005-09-22 Medtronic, Inc. Methods and systems for treatment of neurological diseases of the central nervous system
US20090297592A1 (en) 2005-05-11 2009-12-03 Jcr Pharmaceuticals Co., Ltd. Lipid Vesicle Composition
US20110105560A1 (en) 2005-06-08 2011-05-05 Amicus Therapeutics, Inc. Treatment of cns disorders associated with mutations in genes encoding lysosomal enzymes
US20090130079A1 (en) 2006-01-20 2009-05-21 Genzyme Corporation Intraventricular enzyme delivery for lysosomal storage diseases
US20090246187A1 (en) 2006-04-04 2009-10-01 Shire Pharmaceuticals Ireland Limited Process for concentration of a polypeptide
WO2007141346A2 (en) 2006-06-09 2007-12-13 Novartis Ag Genes regulating intracellular cholesterol trafficking as targets for treatment of cholesterol-related diseases
WO2008070769A1 (en) 2006-12-06 2008-06-12 Medtronic, Inc. Intrathecal catheter
US20090041741A1 (en) 2007-03-06 2009-02-12 Saint Louis University Modified enzyme and treatment method
US20100260706A1 (en) 2007-08-15 2010-10-14 Oren Bogin Compositions and methods for improving production of recombinant polypeptides
US20090291062A1 (en) 2007-11-30 2009-11-26 Wolfgang Fraunhofer Protein formulations and methods of making same
US20100068195A1 (en) 2008-01-18 2010-03-18 Biomarin Pharmaceutical Inc. Manufacture of Active Highly Phosphorylated Human Lysosomal Sulfatase Enzymes and Uses Thereof
WO2009137721A2 (en) 2008-05-07 2009-11-12 Zystor Therapeutics, Inc. Lysosomal targeting peptides and uses thereof
US20120213762A1 (en) 2008-05-07 2012-08-23 Biomarin Pharmaceutical Inc. Lysosomal targeting peptides and uses thereof
US20130168961A1 (en) 2009-06-29 2013-07-04 LighSail Energy Inc. Compressed air energy storage system utilizing two-phase flow to facilitate heat exchange
US20110318327A1 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Treatment of sanfilippo syndrome type b
US8545837B2 (en) 2010-06-25 2013-10-01 Shire Human Genetic Therapies, Inc. Methods and compositions for CNS delivery of iduronate-2-sulfatase
US20110318323A1 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of iduronate-2-sulfatase
WO2011163648A1 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Cns delivery of therapeutic agents
WO2011163647A2 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of heparan n-sulfatase
WO2011163651A2 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. METHODS AND COMPOSITIONS FOR CNS DELIVERY OF β-GALACTOCEREBROSIDASE
US20120003202A1 (en) 2010-06-25 2012-01-05 Shire Human Genetic Therapies, Inc. Cns delivery of therapeutic agents
US20120009171A1 (en) 2010-06-25 2012-01-12 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of arylsulfatase a
US20120014936A1 (en) 2010-06-25 2012-01-19 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of heparan n-sulfatase
US20140271598A1 (en) 2010-06-25 2014-09-18 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of iduronate-2-sulfatase
WO2011163652A2 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Treatment of sanfilippo syndrome type b
US20110318324A1 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of b-galactocerebrosidase
WO2011163649A2 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc Methods and compositions for cns delivery of iduronate-2-sulfatase
WO2011163650A2 (en) 2010-06-25 2011-12-29 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of arylsulfatase a
US20130295077A1 (en) 2010-06-25 2013-11-07 Shire Human Genetic Therapies, Inc. Treatment of sanfilippo syndrome type b
US20130295071A1 (en) 2010-06-25 2013-11-07 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of b-galactocerebrosidase
WO2012023623A2 (ja) 2010-08-19 2012-02-23 日本ケミカルリサーチ株式会社 ハンター症候群の治療剤

Non-Patent Citations (163)

* Cited by examiner, † Cited by third party
Title
Altschul et al., "Basic logic alignment search tool," J. Mol. Biol., 215(3): 403-410, 1990.
Altschul et al., "Gapped BLAST and PSI-BLAST: a new generation of protein database search programs," Nucleic Acids Res. 25: 3389-3402, 1997.
Altschul et al., "Local alignment statistics," 266:460-80, Methods in Enzymology., 1996.
Ammaya et al., "Subcutaneous Reservoir and Pump for Sterile Access to Ventricular Cerebrospinal Fluid," Lancet 2(7315): 983-984, 1963.
Anonymous, TKT to Present Research on Intrathecal Delivery of I2S for Hunter Syndrome at ASHG, PRNewswire, 1 (2004).
Anonymous, TKT's Research Findings on Intrathecal Delivery of I2S Presented at ASHG, Evaluate Ltd, 1 (2004).
Baskin, G. et al., "Genetic galactocerebrosidase deficiency (globoid cell leukodystrophy, Krabbe disease) in rhesus monkeys (Macaca mulatta)," Lab Anim. Sci., 48(5): 476-482, 1998.
Baum, H. et al., "The assay of arylsulphatases A and B in human urine," Clin Chim Acta. 4(3): 453-455, 1959.
Begley et al., "Lysosomal storage diseases and the blood-brain barrier," Curr Pharm Des 14(16): 1566-1580, 2008.
Belichenko et al., Penetration, diffusion, and uptake of recombinant human alpha-L-iduronidase after intraventricular injection into the rat brain, Mol. Genet. Metab., 86(1-2): 141-149, 2005.
Beniaminovitz et al., "Prevention of rejection in cardiac transplantation by blockage of the interleukin-2 receptor with a monoclonal antibody," N. Engl. J. Med. 342(9): 613-619, 2000.
Berard et al., "A review of interleukin-2 receptor antagonists in solid organ transplantation," Pharmacotherapy 19(10): 1127-1137, 1999.
Bielicki et al., "Recombinant human sulphamidase: expression, amplification, purification and characterization," Journal of Biochemistry, 329(Pt 1): 145-150, 1998.
Biswas S. et al., "Substrate reduction intervention by L-cycloserine in twitcher mice (globoid cell leukodystrophy) on a B6; CAST/Ei background," Neurosci. Lett., 347(1): 33-36, 2003.
Blasberg, R.G. et al., "Intrathecal chemotherapy: brain tissue profiles after ventriculocisternal perfusion," J Pharmacol Exp Ther. 195(1): 73-83, 1975.
Bobo et al., "Convection-enhanced delivery of macromolecules in the brain," Proc. Natl. Acad. Sci. U.S.A. 91(6), 2076-2080, 1994.
Bowman, R.H., "Inhibition of citrate metabolism by sodium fluoroacetate in the perfused rat heart and the effect on phosphofructokinase activity and glucose utilization," 93(2): 13C-15C, 1964.
Branco et al., "Selective deletion of antigen-specific, activated T cells by a humanized MAB to CD2 (MEDI-507) is mediated by NK cells," Transplantation 68(10): 1588-1596, 1999.
Burrow, T. Andrew and Leslie, Nancy D., Review of the use of idursulfase in the treatment of mucopolysaccharidosis II, Biologics: Targets and Therapy, 2(2):311-320 (2008).
Butt MT, "Morphologic changes associated with intrathecal catheters for direct delivery to the central nervous system im preclinical studies," Toxicol. Pathol., 39(1): 213-219, 2011.
Cabrera-Salazar, M.A. et al., "Intracerebroventricular delivery of glucocerebrosidase reduces substrates and increases lifespan in a mouse model of neuronopathic Gaucher disease," Exp Neurol. 225(2): 436-444, 2010.
Champion K. J. et al., Identification and characterization of a novel homozygous deletion in the x-N-acetylglucosaminidase gene in a patient with Sanfilippo type B syndrome (mucopolysaccharidosis IIIB), Molecular Genetics and Metabolism, 100: 51-56 (2010).
Chirmule et al., "Readministration of adenovirus vector in nonhuman primate lungs by blockage of CD40-CD40 ligand interactions," J. Virol. 74(7): 3345-3352, 2000.
Chiro et al., "Spinal descent of cerebrospinal fluid in man," Neurology 26(1): 1-8, 1976.
Clarke, L. A., Idursulfase for the treatment of mucopolysaccharidosis II, Expert Opin. Pharmacother., 9(2):311-317 (2008).
Cressent, A. et al., Improved Behavior and Neuropathology in the Mouse Model of Sanfilippo Type IIIB Disease after Adeno-Associated Virus-Mediated Gene Transfer in the Striatum, The Journal of Neuroscience, 24(45): 10229-10239 (2004).
Dekaban AS., "Changes in brain weights during the span of human life: relation of brain weights to body heights and body weights," Ann Neurol 4: 345-356, 1978.
Descartes, M. et al., Enzyme Replacement Therapy for MPS II: Developing a Pre-Medication Protocol, University of Alabama and Children's Hospital of Alabama, 1 (2007).
Desnick, R.J., "Enzyme replacement and enhancement therapies for lysosomal diseases," J. Inherit. Metab. Dis., 27(3): 385-410, 2004.
Dickson, P. et al., Intrathecal enzyme replacement therapy: Successful treatment of brain disease via the cerebrospinal fluid, Molecular Genetics & Metabolism, 91(1):61-68 (2007).
Dickson, P.I., Novel Treatments and Future Perspectives: Outcomes of Intrathecal Drug Delivery, International Journal of Clinical Pharmacology and Therapeutics, 47:1 S124-127 (2009).
Eckhoff et al., "The safety and efficacy of a two-dose daclizumab (zenapax) induction therapy in liver transplant recipients," Transplantation 69(9): 1867-1872, 2000.
Ekberg et al., "Daclizumab prevents acute rejection and improves patient survival post transplantation: 1 year pooled analysis," Transpl. Int. 13(2): 151-159, 2000.
Elaprase (idursulfase), http://www.elaprase.com/pdf/Elaprase—Overview—Sheet110811.pdf, REV 5, 2011.
Elaprase idursulface, European Medicines Agency-Science, Medicines, Health, XP-002716697, pp. 1-3 (2007).
EMEA: "Elaprase - Idursulfase", EUROPEAN MEDICINES AGENCY - FIND MEDICINE - ELAPRASE, EUROPEAN MEDICINES AGENCY - SCIENCE, MEDICINES, HEALTH, pages 1 - 3, XP002716697, Retrieved from the Internet <URL:www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines> [retrieved on 20131120]
Esposito, S. et al, Heparan N-sulfatase gene: two novel mutations and transient expression of 15 defects, Biochimica et Biophysica Acta 1501, 1-11: 1 (2000).
European Search Report for EP, 11799034.1, 8 pages (dated Mar. 12, 2014).
Extended European Search Report for 11799039.0, 12 pages (dated Jun. 10, 2014).
Extended European Search Report for EP11799035.8, 7 pages, dated Dec. 16, 2013.
Felice, B.R. et al., Safety Evaluation of Chronic Intrathecal Administration of Idursulfase-IT in Cynomolgus Monkeys, Toxicology Pathology, 39:879-892 (2011).
Fenstermacher et al., "Drug "diffusion" within the brain," Ann NY Acad Sci 531: 29-39, 1988.
Ficko-Blean E, et al., "Structural and mechanistic insight into the basis of mucopolysaccharidosis IIIB," PNAS, 105(18): 6560-6565, 2008.
Fishwild et al., "Differential effects of administration of a human anti-CD4 monoclonal antibody, HM6G, in nonhuman primates," Clin. Immunol. 92(2): 138-152, 1999.
Fu, H. et al., Restoration of Central Nervous System a-N-Acetylglucosaminidase Activity and Therapeutic Benefits in Mucopolysaccharidosis IIIB Mice by a Single Intracisternal Recombinant Adeno-Associated Viral Type 2 Vector Delivery, The Journal of Gene Medicine, 12:624-633 (2010).
Fu, H. et al., Significantly Increased Lifespan and Improved Behavioral Performances by rAAV Gene Delivery in Adult Mucopolysaccharidosis IIIB Mice, Gene Therapy 14:1065-1077 (2007).
Garbuzova-Davis, S. et al., Transplantation of Human Umbilical Cord Blood Cells Benefits an Animal Model of Sanfilippo Syndrome Type B, Stem Cells and Development, 14:384-394 (2005).
Garcia, A.R. et al., Intrathecal Delivery of Iduronate 2-Sulfatase to the CNS of Cynomolgous Monkeys, Shire Human Genetic Therapies, 1 (2007).
Gaziev et al., "Chronic graft-versus-host disease: is there an alternative to the conventional treatment?," Bone Marrow Transplant, 25(7): 689-696, 2000.
GenBank accession No. NM000263, Homo sapiens N-Acetylglucosaminidase, Alpha (NAGLU) mRNA, 1-4 (accessed May 3, 2014).
GeneCards, Galactosylceramidase, http://www.genecards.org/cgi-bin/carddisp.pl?gene=GALC&search=Galactocerebrosidase, 2012.
Ghersi-Egea, J.F. et al, "Rapid distribution of intraventricularly administered sucrose into cerebrospinal fluid cisterns via subarachnoid velae in rat," Neuroscience 75(4): 1271-1288, 1996.
Graham et al., "Characteristics of a human cell line transformed by DNA from human adenovirus type 5," J. Gen Virol., 36(1): 59-74, 1977.
Greene, et al., Metachromatic Leukodystriphym, Arch. Neurol. , vol. 20, Feb. 1969, pp. 147-153.
GREMPE R., SARETZ B.: "MATERIAL IN DER MONTAGE AUFTRAGSBEZOGEN BEREITSTELLEN.", ZWF ZEITSCHRIFT FUR WIRTSCHAFTLICHE FERTIGUNG UNDAUTOMATISIERUNG., CARL HANSER VERLAG. MUNCHEN., DE, vol. 87., no. 01., 1 January 1992 (1992-01-01), DE, pages 51 - 54., XP000271916, ISSN: 0947-0085
Grubb JH et al., "New strategies for enzyme replacement therapy for lysosomal storage diseases," Rejuvenation Research 13(2-3): 229-236, 2010.
Gummert et al., "Newer immunosuppressive drugs: a review," J. Am. Soc. Nephrol, 10(6): 1366-1380, 1999.
Hashimoto R, "N-terminal deletion mutants of insulin-like growth factor-II (IGF-II) show Thr7 and Leu8 important for binding to insulin and IGF-I receptors and Leu8 critical for all IGF-II functions," J Biol Chem., 270(30); 18013-18018, 1995.
Hemsley, et al., Effect of high dose, repeated intra-cerebrospinal fluid injection of sulphamidase on neuropathology in mucopolysaccharidosis type IIIA mise, Genes, Brain and Behavior, 7:740-753, (2008), pp. 740-753.
Hemsley, Kim M. et al., "Injection of recombinant human sulfamidase into the CSF via the cerebellomedullary cistern in MPS IIIA mice," Mol Genet Metab. 90(3): 313-328, 2007.
Henry ML, "Cyclosporine and tacrolimus (FK506): a comparison of efficacy and safety profiles," Clin. Transplant, 13(3): 209-220, 1999.
Hong et al., "Immunosuppressive agents in organ transplantation: past, present, and future," Semin. Nephrol. 20(2): 108-125, 2000.
Hood RD, Development and Reproductive Toxicology: A practical approach, 276, 2006.
Hoogerbrugge, P.M., et al., "Effect of bone marrow transplantation on enzyme levels and clinical course in the neurologically affected," J. Clin. Invest., 81(6): 1790-1794, 1988.
Hovland DN, et al., "Six-month continuous intraputamenal infusion toxicity study of recombinant methionyl human glial cell line-derived neurotrophic factor (r-metHuGDNF in rhesus monkeys," Toxicol. Pathol., 35(7): 1013-1029, 2007.
Ideguchi et al., "Local adenovirus-mediated CFLA40immunoglobulin expression suppresses the immune responses to adenovirus vectors in the brain," Neuroscience 95(1): 217-226, 2000.
International Preliminary Report on Patentability for PCT/US11/41928, 36 pages (dated Mar. 29, 2013).
International Search Report for PCT/US11/41922, dated Feb. 14, 2012.
International Search Report for PCT/US11/41924, dated Nov. 7, 2011.
International Search Report for PCT/US11/41925, dated Feb. 14, 2012.
International Search Report for PCT/US11/41926, 5 pages (dated May 13, 2013).
International Search Report for PCT/US11/41927, dated Mar. 9, 2012.
International Search Report for PCT/US2011/041928,4 pages (dated Sep. 26, 2012).
Ito et al., "Induction of CTL responses by simultaneous administration of liposomal peptide vaccine with anti-CD40 and anti-CTLA-4 mAb," J. Immunol. 164(3): 1230-1235, 2000.
Johanson CE, et al., "Multiplicity of cerebrospinal fluid functions: New challenges in health and disease," Cerebrospinal Fluid Res., 14(5): 10, 2008.
Johnson, K., "Globoid leukodystrophy in the cat," J. Am. Vet. Med. Assoc., 157(12): 2057-2064, 1970.
Joshi S. et al., "Targeting the brain: rationalizing the novel methods of drug delivery to the central nervous system," Neurocrit Care 6(3): 200-212, 2007.
Kakkis et al., Mol. Genet. and Metab. 83: 163-174 (2004). *
Kakkis, E. et al., Intrathecal enzyme replacement therapy reduces lysosomal storage in the brain and meninges of the canine model of MPS I, Molecular Genetics and Metabolism 83:163-174 (2004).
Kang, H. et al., Significantly increased lifespan and improved behavioral performances by rAAV gene delivery in adult mucupolysaccharidosis IIIB mice, Gene Therapy, 14:1066-1077 (2007).
Kerwin, J. Pharm. Sci. 97: 2924-2935 (2008). *
Kobayashi T. et al., "The Twitcher mouse: an enzymatically authentic model of human globoid cell leukodystrophy (Krabbe disease)," Brain Res., 202(2): 479-483, 1980.
Krewson, CE et al., "Distribution of nerve growth factor following direct delivery to brain interstitium," Brain Res. 680(1-2): 196-206, 1995.
Kurlberg et al., "Blockage of the B7-CD28 pathway by CTLA4-lg counteracts rejection and prolongs survival in small bowel transplantation," Scand. J. Immunol, 51(3): 224-230, 2000.
Lamsa, J.C et al., Delivery of I2S to the Canine CNS: Comparison of Intracisternal, Intralumbar and Intraventricular Dose Routes as Potential Treatents for Severe MPS II, Shire HGT, 1 (2006).
Lamsa, J.C. et al., Intrathecal Delivery of Iduronate 2-Sulfatase for MPS II to the Canine CNS, ASHG Annual Meeting, 1 (2004).
Lazorthes et al., Advances in Drug Delivery Systems and Application in Neurosurgery, 18: 143-192, 1991.
Lee, et al., "Single-dose intracerebroventricular administration of galactocerebrosidase improves survival in a mouse model of globoid cell leukodystrophy," FASEB Journal, 21(10): 2520-2527, 2007.
LeVine S. et al., "L-cycloserine slows the clinical and pathological course in mice with globoid cell leukodystrophy (twitcher mice)," J. Neurosci. Res., 60(2): 231-236, 2000.
Li HH, et al., "Mouse model of Sanfilippo syndrome type B produced by targeted disruption of the gene encoding alpha-N-acetylglucosaminidase," PNAS 96(25): 14505-14510, 1999.
Li, et al., "Attenuated plasticity in neurons and astrocytes in the mouse model of Sanfilippo syndrome type B," J Neurosci Res, 69(1): 30-8, 2002.
Lin, D., et al., "Central nervous system-directed AAV2/5-mediated gene therapy synergizes with bone marrow transplantation in the murine model of globoid-cell leukodystrophy," Mol. Ther., 15(1): 44-52, 2007.
Lu, Y. et al., Direct Brain Delivery of Iduronate 2-Sulfastase Reduces Glycosaminoglycan Accumulation and Improves Histopathology in the CNS and Peripheral Tissue of Hunter Mice, Shire HGT, 1 (2007).
Luca, Tonia, "Axons mediate the distribution of arylsulfatase A within the mouse hippocampus upon gene delivery," Mol Ther. 12(4): 669-679, 2005.
Marinova-Mutafchieva et al., "A comparative study into the mechanisms of action of anti-tumor necrosis factor alpha, anti-CD4, and combined anti-tumor necrosis factor alpha/anti-CD4 treatment in early collagen-induced arthritis," Arthritis Rheum 43: 638-644, 2000.
Mather et al., "Culture of testicular cells in hormone-supplemented serum-free medium," Ann N.Y. Acad. Sci., 383: 44-68, 1982.
Mather, "Establishment and characterization of two distinct mouse testicular epithelial cell lines," Biol. Reprod., 23: 243-251, 1980.
Matheus, MG et al., "Brain MRI findings in patients with mucopolysaccharidosis types I and Ii and mild clinical presentation," Neuroradiology 46(8): 666-672, 2004.
Matzner et al., Hum. Mol. Gen. 14(9): 1139-1152 (2005). *
Meikle et al., "Diagnosis of lysosomal storage disorders: evaluation of lysosome-associated membrane protein LAMP-1 as a diagnostic marker," Clin Chem., 43(8 Pt 1): 1325-1335, 1997.
Middaugh et al., "Determination of the apparent thermodynamic activities of saturated protein solutions," J. Biol. Chem. 254(2): 367-370, 1979.
Moder, KG., "New medications for use in patients with rheumatoid arthritis," Ann. Allergy. Asthma Immunol. 84(3): 280-284, 2000.
Nagaraja, TN et al., "In normal rat, intraventricularly administered insulin-like growth factor-1 is rapidly cleared from CSF with limited distribution into brain," Cerebrospinal Fluid Res. 2: 1-15, 2005.
Nail S.L. et al., "Fundamentals of freeze-drying, in Development and manufacture of protein pharmaceuticals," Nail S.L. editor New York: Kluwer Academic/Plenum Publishers, 281-353, 2002.
Neufeld EF, Muenzer J., "The mucopolysaccharidoses," In: Scriver CR, Beaudet AI, Sly WS, et al, eds. The Metabolic and Molecular Bases of Inherited Disease. www.ommbid.com 8th ed. New York, NY: McGraw-Hill; 2001:3421-3452.
Neufeld, E.F., Enzyme Replacement therapy. Lysosomal disorders of the Brain, ed. F.M.a.W. Platt, S.V. 2004: Oxford University Press: 327-338, 2004.
Nevins, TE., "Overview of new immunosuppressive therapies," Curr. Opin. Pediatr. 12(2): 146-150, 2000.
Nguyen et al., "Convective distribution of macromolecules in the primate brain demonstrated using computerized tomography and magnetic resonance imaging," J. Neurosurg. 98(3), 584-590, 2003.
Ohmi, et al., "Activated microglia in cortex of mouse models of mucopolysaccharidoses I and IIIB," Proc Natl Acad Sci, 100(4): 1902-7, 2002.
Okuyama, T. et al., Japan Elaprase® Treatment (JET) study: Idursulfase enzyme replacement therapy in adult patients with attenuated Hunter syndrome (Mucopolysaccharidosis II, MPS II), Molecular Genetics and Metabolism, 99:18-25 (2010).
Ommaya et al., "Implantable devices for chronic access and drug delivery to the central nervous system," Cancer Drug Delivery, 1(2): 169-179, 1984.
Pardridge WM., "Drug transport in brain via the cerebrospinal fluid," Fluids Barriers CNS, 8(1): 7, 2011.
Passini, MA et al., "Distribution of a lysosomal enzyme in the adult brain by axonal transport and by cells of the rostral migratory stream," J Neurosci 22(15): 6437-6446, 2002.
Penn, RD et al., "Intrathecal ciliary neurotrophic factor delivery for treatment of amyotrophic lateral sclerosis (phase I trial)," Neurosurgery 40(1): 94-99, 1997.
Phosphate Buffer Calculation, http://www.egr.msu.edu/biofuelcell/tools/phosphate/phosphate.html, Dec. 31, 2000, accessed Aug. 28, 2012.
Ponce RP, et al., "Immunogenicity of biologically-derived therapeutics: assessment and interpretation of nonclinical safety studies," Regul. Toxicol. Pharmacol., 54(2): 164-182, 2009.
Ponticelli et al., "Promising new agents in the prevention of transplant rejection," Drugs R.D. 1(1), 55-60, 1999.
Potter et al., "Review—the use of immunosuppressive agents to prevent neutralizing antibodies against a transgene product," Ann. N.Y. Acad. Sci. 875: 159-174, 1999.
Pritchard, D. et al., "Globoid cell leucodystrophy in polled Dorset sheet," Vet. Pathol., 17(4): 399-405, 1980.
Przepiorka et al., "A phase II study of BTI-322, a monoclonal anti-CD2 antibody, for treatment of steroid-resistant acute graft-versus-host disease," Blood 92(11): 4066-4071, 1998.
Qi et al., "Effect of tacrolimus (FK506) and sirolimus (rapamycin) mono- and combination therapy in prolongation of renal allograft survival in the monkey," Transplantation 69(7), 1275-1283, 2000.
Rieselbach RE et al., "Subarachnoid distribution of drugs after lumbar injection," N Engl J Med. 267(25): 1273-1278, 1962.
Savas, et al., "Intracerebral injection of sulfamidase delays neuropathology in murine MPS-IIIA," Mol Genet Metab., 82(4): 273-285, 2004.
Schlessingerman, A., Mass of an Adult, obtained from hypertextbook.com/facts/2003/AlexSchlessingerman.shtml, 2003, 2 pages.
Scientific Discussion-Elaprase, XP00271916, pp. 1-43 (2007).
Shahrokh et al., "Intrathecal delivery of protein therapeutics to treat genetic diseases involving the CNS, in: Injectable Drug Delivery 2010: Formulations Focus," ONdrugDelivery, pp. 16-20, 2010.
Shire Human Genetic Therapies, Intrathecal Delivery of Protein Therapeutics to Treat Genetic Diseases Involving the CNS, www.ondrugdelivery.com, pp. 16-20, (Publically available on Jun. 30, 2010).
Simard JM et al., "Brain oedema in focal ischaemia: molecular pathophysiology and theoretical implications," Lancet Neurol. 6(3): 258-268, 2007.
Sinow, C.S., Construction of an IGF-NAGLU Fusion Protein for Treatment of Sanfilippo B Syndrome, California State Sciene Fair, 1 (2008).
Sjoberg, M. et al., Long-term Intrathecal Morphine and Bupivacaine in Patients with Refractory Cancer Pain, Anesthesiology, 80:284-297 (1994).
Slavik et al., "CD28/CTLA-4 and CD80/CD86 families: signaling and function," Immunol Res. 19(1): 1-24, 1999.
Stamatovic SM, et al., "Brain endothelial cell-cell junctions: how to "open" the blood brain barrier," Curr. Neuropharmacol., 6(3): 179-192, 2008.
Stroobants S. et al., "Intracerebroventricular enzyme infusion corrects central nervous system pathology and dysfunction in a mouse model of metachromatic leukodystrophy," Hum Mol Genet. 20(14): 2760-2769, 2011.
Sturk, et al., "Combined Intracerebroventricular Intraperitoneal Enzyme Replacement Therapy Improves Survival and Reduces Brain Psychosine in a Mouse Model of Krabbe Disease," European Task Force on Brain and Neurogenerative Lysosomal Storage Diseases, http://www.brains4brain.eu/assets/files/abstract-francoforte-2009.pdf p. 42, 2009.
Tang, X. et al., "Design of freeze-drying processes for pharmaceuticals: Practical advice," Pharm. Res., 21(2): 191-200, 2004.
Tippin, B. et al., Insulin-like Growth Factor-2 Peptide Fusion Enables Uptake and Lysosomal Delivery of N-Acetylglucosamindidase to Mucopolysaccharidosis IIIB Fibrboblasts, MPS Scientific Program: Plenary Papers, entire document: p. 100 (Jun. 26, 2010).
Toyoshima, E. et al., "Nerve conduction studies in the Twitcher mouse (murine globoid cell leukodystrophy)," J. Neurol. Sci., 74(2-3): 307-318, 1986.
Urlaub and Chasin, "Isolation of Chinese hamster cell mutants deficient in dihydrofolate reductase activity," Proc. Natl. Acad. Sci. USA, 77(7): 4216-4220, 1980.
Vedolin, L. et al., "Correlation of MR imaging and MR spectroscopy findings with cognitive impairment in mucopolysaccharidosis II," AJNR Am J Neuroradial 28(6): 1029-1033, 2007.
Vertemati, T. et al., Multidisciplinary Evaluation in 12 Mucopolysaccharidose Type II or Hunter Syndrome Patients Prior Enzyme Replacement Therapy, CREIM, UNIFESP, 1 (2007).
Vite, Charles H. et al., "Biodistribution and pharmacodynamics of recombinant human alpha-L-iduronidase (rhIDU) in mucopolysaccharidosis type I-affected cats following multiple intrathecal administrations," Mol Genet Metab 103(3): 268-274, 2011.
Vogler, C. et al., "Overcoming the blood-brain barrier with high-dose enzyme replacement therapy in murine mucopolysaccharidosis VII," Proc Natl Acad Sci USA 102(41): 14777-14782, 2005.
Waheed, A et al., "Purification of mammalian arylsulfatase A enzymes by subunit affinity chromatography," Int J Pept Protein Res., 26(4): 362-372, 1985.
Walkley, "Cell Pathology of lysosomal storage disorders," Brain Pathol., 8, 175-93, 1998.
Wang et al., "Lyophilization and development of solid protein pharmaceuticals," Int. J. Pharm., 203(1-2): 1-60, 2000.
Wang et al., "Treatment reduces or stabilizes brain imaging abnormalities in patients with MPS I and II," Molecular Genetics and Metabolism, 98(4): 406-11, 2009.
Wang, W. and Roberts, C., Aggregation of Therapeutic Proteins, published by John Wiley & Sons, Inc., Hoboken, New Jersey (2010).
Watson et al., "Intrathecal administration of AAV vectors for the treatment of lysosomal storage in the brains of MPS I mice," Gene Ther., 13(11): 917-925, 2006.
Weber, B. et al., Novel Mutations in Sanfilippo A syndrome: Implications for Enzyme function, Hum. Mol. Genet., 6(9): 1573-1579 (1997).
Wenger, D.A. et al., Galactosylceramide Lipidosis: Globoid Cell Leukodystrophy (Krabbe Disease), in the Metabolic and Molecular Bases of Inherited Disease, C.R. Scriver, Beaudet, A., Sly, W.S. and Valle, D. Editor 2001 McGraw-Hill, 3669-3687, 2001.
Wenger, D.A., "Murine, canine and non-human primate models of Krabbe disease," Mol. Med. Today, 6(11): 449-451, 2000.
Williams N. A. et al., "The lyophilization of pharmaceuticals; A literature review." J. Parenter Sci. Technol., 38(2): 48-59, 1984.
Wiseman et al., "Daclizumab: a review of its use in the prevention of acute rejection in renal transplant recipients," Drugs 58(6): 1029-1042, 1999.
Won, C., Stabilizers against heat-induced aggregation of RPR 114849, an acidic fibroblast growth factor (aFGF), International Journal of Pharmaceutics, 167:25-36 (1998).
Wraith, J.E. et al., Mucopolysaccharidosis type II (Hunter syndrome): a clinical review and recommendations for treatment in the era of enzyme replacement therapy, Eur. J. Pediatr., 167: 247-277 (2008).
Written Opinion for PCT/US11/41922, dated Feb. 14, 2012.
Written Opinion for PCT/US11/41924, dated Nov. 7, 2011.
Written Opinion for PCT/US11/41925, dated Feb. 14, 2012.
Written Opinion for PCT/US11/41926, 8 pages (dated May 13, 2013).
Written Opinion for PCT/US11/41927, dated Mar. 9, 2012.
Written Opinion for PCT/US2011/041928,13 pages (dated Sep. 26, 2012).
Yan Q et al., "Distribution of intracerebral ventricularly administered neurotrophins in rat brain and its correlation with trk receptor expression," Exp Neurol. 127(1): 23-36, 1994.
Yeager A. et al., "Prolonged survival and remyelination after hematopoietic cell transplantation in the twitcher mouse," Science, 225(4666): 1052-1054, 1984.

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11471516B2 (en) * 2010-06-25 2022-10-18 Shire Human Genetic Therapies, Inc. CNS delivery of therapeutic agents
US20180036388A1 (en) * 2013-05-15 2018-02-08 Regenxbio Inc. Adeno-associated virus mediated gene transfer to the central nervous system
US20180099030A1 (en) * 2013-05-15 2018-04-12 Regents Of The University Of Minnesota Methods to treat mucopolysaccharide type ii or deficiency in iduronate-2-sulfatase using a recombinant adeno-associated virus encoding iduronate-2-sulfatase
US11020461B2 (en) * 2016-02-17 2021-06-01 Takeda Pharmaceutical Company Limited Methods and compositions for CNS delivery of arylsulfatase A
US11819539B2 (en) 2017-09-22 2023-11-21 The Trustees Of The University Of Pennsylvania Gene therapy for treating Mucopolysaccharidosis type II
WO2021061517A1 (en) * 2019-09-23 2021-04-01 University Of Washington Assay of arylsulfatase a enzymatic activity in dried blood spots by mass spectrometry and fluorometry

Also Published As

Publication number Publication date
NZ605873A (en) 2015-02-27
KR102537084B1 (ko) 2023-05-26
AU2021236557A1 (en) 2021-10-28
KR20180059580A (ko) 2018-06-04
JP6045493B2 (ja) 2016-12-14
HRP20191923T1 (hr) 2020-01-10
AU2017202289A1 (en) 2017-04-27
KR102318997B1 (ko) 2021-10-29
KR20130135821A (ko) 2013-12-11
EP2585104A4 (en) 2014-01-15
KR102094094B1 (ko) 2020-03-27
RU2671503C2 (ru) 2018-11-01
WO2011163650A2 (en) 2011-12-29
PL2585104T3 (pl) 2020-01-31
AU2019204051B2 (en) 2021-07-22
LT2585104T (lt) 2019-10-25
AU2019204051A1 (en) 2019-08-29
KR20200035167A (ko) 2020-04-01
AU2017202289B2 (en) 2019-03-14
TW201206464A (en) 2012-02-16
CA2803003C (en) 2022-11-22
KR20230079472A (ko) 2023-06-07
RU2012154574A (ru) 2014-07-27
RS59468B1 (sr) 2019-11-29
MX2013000323A (es) 2013-06-05
KR20210131449A (ko) 2021-11-02
PL3626257T3 (pl) 2022-03-28
BR112012033216B1 (pt) 2022-10-11
IL291556B2 (en) 2024-02-01
WO2011163650A3 (en) 2013-06-13
EP3626257A1 (en) 2020-03-25
KR101997935B1 (ko) 2019-10-04
AU2011270670B2 (en) 2017-01-12
IL273854B (en) 2022-04-01
IL273854A (en) 2020-05-31
MX344795B (es) 2017-01-05
SI2585104T1 (sl) 2019-11-29
EP3626257B1 (en) 2021-08-04
JP2013538788A (ja) 2013-10-17
DK2585104T3 (da) 2019-10-14
NZ702799A (en) 2016-08-26
US20120009171A1 (en) 2012-01-12
IL223758B (en) 2020-04-30
BR112012033216A2 (pt) 2020-08-25
ZA201300675B (en) 2017-11-29
CY1124910T1 (el) 2023-01-05
IL291556B1 (en) 2023-10-01
UA115650C2 (uk) 2017-12-11
EP2585104B1 (en) 2019-08-21
CY1122282T1 (el) 2020-11-25
EP3964230A1 (en) 2022-03-09
TWI667037B (zh) 2019-08-01
IL291556A (en) 2022-05-01
CN103282046A (zh) 2013-09-04
CA2803003A1 (en) 2011-12-29
EP2585104A2 (en) 2013-05-01
AU2011270670A1 (en) 2013-02-14
ES2754776T3 (es) 2020-04-20
ME03647B (me) 2020-07-20
CN103282046B (zh) 2015-05-13

Similar Documents

Publication Publication Date Title
US20200405825A1 (en) Methods and Compositions for CNS Delivery of Arylsulfatase A
AU2019204051B2 (en) Methods and compositions for cns delivery of arylsulfatase a
AU2021202579B2 (en) Methods and compositions for cns delivery of iduronate-2-sulfatase
US20200237883A1 (en) Stable formulations for cns delivery of arylsulfatase a
US20130295071A1 (en) Methods and compositions for cns delivery of b-galactocerebrosidase
RU2783380C2 (ru) Способы и композиции для доставки в цнс арилсульфатазы а

Legal Events

Date Code Title Description
AS Assignment

Owner name: SHIRE HUMAN GENETIC THERAPIES, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SALAMAT-MILLER, NAZILA;TAYLOR, KATHERINE;CAMPOLIETO, PAUL;AND OTHERS;SIGNING DATES FROM 20110805 TO 20110817;REEL/FRAME:026825/0460

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

AS Assignment

Owner name: TAKEDA PHARMACEUTICAL COMPANY LIMITED, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SHIRE HUMAN GENETIC THERAPIES, INC.;REEL/FRAME:061447/0760

Effective date: 20220711